EP3693475A1 - Methoden und materialien zur beurteilung eines homologen rekombinationsfehlers - Google Patents

Methoden und materialien zur beurteilung eines homologen rekombinationsfehlers Download PDF

Info

Publication number
EP3693475A1
EP3693475A1 EP19216740.1A EP19216740A EP3693475A1 EP 3693475 A1 EP3693475 A1 EP 3693475A1 EP 19216740 A EP19216740 A EP 19216740A EP 3693475 A1 EP3693475 A1 EP 3693475A1
Authority
EP
European Patent Office
Prior art keywords
regions
indicator
loh
tai
hrd
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP19216740.1A
Other languages
English (en)
French (fr)
Inventor
Victor Abkevich
Kirsten Timms
Alexander Gutin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Myriad Genetics Inc
Original Assignee
Myriad Genetics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=51659359&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=EP3693475(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Myriad Genetics Inc filed Critical Myriad Genetics Inc
Publication of EP3693475A1 publication Critical patent/EP3693475A1/de
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6813Hybridisation assays
    • C12Q1/6827Hybridisation assays for detection of mutation or polymorphism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6844Nucleic acid amplification reactions
    • C12Q1/6858Allele-specific amplification
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations
    • G16B20/20Allele or variant detection, e.g. single nucleotide polymorphism [SNP] detection
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/154Methylation markers
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers
    • GPHYSICS
    • G16INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR SPECIFIC APPLICATION FIELDS
    • G16BBIOINFORMATICS, i.e. INFORMATION AND COMMUNICATION TECHNOLOGY [ICT] SPECIALLY ADAPTED FOR GENETIC OR PROTEIN-RELATED DATA PROCESSING IN COMPUTATIONAL MOLECULAR BIOLOGY
    • G16B20/00ICT specially adapted for functional genomics or proteomics, e.g. genotype-phenotype associations

Definitions

  • This document relates to methods and materials involved in assessing samples (e.g., cancer cells or nucleic acids derived therefrom) for homologous recombination deficiency (HRD) (e.g., an HRD signature) based on detection of particular chromosomal aberrations ("CA").
  • HRD homologous recombination deficiency
  • CA chromosomal aberrations
  • this document provides methods and materials for detecting CA Regions to determine whether or not a cell (e.g., a cancer cell) has HRD (e.g., exhibits an HRD signature).
  • HRD homology-dependent repair
  • Cancer is a serious public health problem, with 562,340 people in the United States of America dying of cancer in 2009 alone.
  • American Cancer Society, Cancer Facts & Figures 2009 (available at American Cancer Society website ).
  • One of the primary challenges in cancer treatment is discovering relevant, clinically useful characteristics of a patient's own cancer and then, based on these characteristics, administering a treatment plan best suited to the patient's cancer. While strides have been made in this field of personalized medicine, there is still a significant need for better molecular diagnostic tools to characterize patients' cancers.
  • one aspect of this invention features a method for assessing HRD in a cancer cell or DNA (e.g. , genomic DNA) derived therefrom.
  • the method comprises, or consists essentially of, (a) detecting, in a sample or DNA derived therefrom, CA Regions (as defined herein) in at least one pair of human chromosomes of sample or DNA derived therefrom ( e.g. , any pair of human chromosomes other than a human X/Y sex chromosome pair); and (b) determining the number, size ( e.g., length), and/or character of said CA Regions.
  • CA Regions are analyzed in a number of chromosome pairs that are representative of the entire genome (e.g. , enough chromosomes are analyzed such that the number and size of CA Regions are expected to be representative of the number and size of CA Regions across the genome).
  • CA Regions useful in such methods include (1) chromosomal regions showing loss of heterozygosity ("LOH Regions", as defined herein), (2) chromosomal regions showing telomeric allelic imbalance (“TAI Regions”, as defined herein), and (3) chromosomal regions showing large scale transition (“LST Regions”, as defined herein).
  • LHO Regions loss of heterozygosity
  • TAI Regions telomeric allelic imbalance
  • LST Regions large scale transition
  • CA Regions of a certain size, chromosomal location or character can be particularly useful in the various aspects of the invention described herein.
  • the invention provides a method of assessing (e.g. , detecting) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of TAI Regions of a certain size or character (e.g. , "Indicator TAI Regions", as defined herein) in the sample; and (3) assessing HRD in the sample based at least in part on the determinations made in (1) and (2).
  • determining the total number of LOH Regions of a certain size or character e.g. , "Indicator LOH Regions", as defined herein
  • TAI Regions e.g. , "Indicator TAI Regions"
  • the invention provides a method of assessing (e.g., detecting) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of LST Regions of a certain size or character (e.g., "Indicator LST Regions", as defined herein) in the sample; and (3) assessing HRD in the sample based at least in part on the determinations made in (1) and (2).
  • the invention provides a method of assessing (e.g., detecting) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of LST Regions of a certain size or character (e.g., "Indicator LST Regions", as defined herein) in the sample; and (3) assessing HR
  • the invention provides a method of assessing ( e.g. , detecting) HRD in a sample comprising (1) determining the total number of TAI Regions of a certain size or character (e.g. , "Indicator TAI Regions", as defined herein) in the sample; (2) determining the total number of LST Regions of a certain size or character ( e.g. , "Indicator LST Regions", as defined herein) in the sample; and (3) assessing HRD in the sample based at least in part on the determinations made in (1) and (2).
  • the invention provides a method of assessing ( e.g. , detecting) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g.
  • the invention provides a method of diagnosing the presence or absence of HRD in a patient sample, the method comprising (1) analyzing (e.g. , assaying) one or more patient samples to determine ( e.g., detect) the total number of LOH Regions of a certain size or character (e.g., "Indicator LOH Regions", as defined herein) in the sample; (2) analyzing ( e.g., assaying) one or more patient samples to determine ( e.g., detect) the total number of TAI Regions of a certain size or character (e.g.
  • the invention provides a method of diagnosing the presence or absence of HRD in a patient sample, the method comprising (1) analyzing (e.g.
  • assaying one or more patient samples to determine ( e.g., detect) the total number of LOH Regions of a certain size or character (e.g., "Indicator LOH Regions", as defined herein) in the sample; (2) analyzing ( e.g. , assaying) one or more patient samples to determine ( e.g. , detect) the total number of LST Regions of a certain size or character (e.g.
  • the invention provides a method of diagnosing the presence or absence of HRD in a patient sample, the method comprising (1) analyzing ( e.g., assaying) one or more patient samples to determine ( e.g., detect) the total number of TAI Regions of a certain size or character ( e.g.
  • the invention provides a method of diagnosing the presence or absence of HRD in a patient sample, the method comprising (1) analyzing (e.g., assaying) one or more patient samples to determine ( e.g., detect) the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) analyzing ( e.g., assaying) one or more patient samples to determine ( e.g., detect) the total number of TAI Regions of a certain size or character (e.g.
  • CA Regions chromosomal regions showing loss of heterozygosity
  • TAI Regions chromosomal regions showing telomeric allelic imbalance
  • LST Regions chromosomal regions showing large scale transition
  • the invention provides a method of assessing (e.g. , detecting) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of TAI Regions of a certain size or character (e.g.
  • assessing (e.g. , detecting) HRD is based on a score derived or calculated from ( e.g. , representing or corresponding to) the detected CA Regions ("CA Region Score", as defined herein). Scores are described in greater detail herein.
  • HRD is detected if a CA Region Score for a sample exceeds some threshold (e.g., a reference or index CA Region Score), and optionally HRD is not detected if the CA Region Score for the sample does not exceed some threshold (e.g., a reference or index CA Region Score, which may in some embodiments be the same threshold for positive detection).
  • some threshold e.g., a reference or index CA Region Score
  • HRD is detected if the CA region Score is below a certain threshold and not detected if the score is above a certain threshold.
  • the CA Region Score is a combination of scores derived or calculated from ( e.g. , representing or corresponding to) two or more of (1) the detected LOH Regions ("LOH Region Score", as defined herein), (2) the detected TAI Regions ("TAI Region Score", as defined herein), and/or (3) the detected LST Regions ("LST Region Score", as defined herein).
  • CA Region Score 0.32 * LOH Region Score + 0.68 * TAI Region Score
  • CA Region Score A * LOH Region Score + B * LST Region Score
  • CA Region Score A * TAI Region Score + B * LST Region Score
  • CA Region Score A * LOH Region Score + B * TAI Region Score + C * LST Region Score
  • CA Region Score 0.21 * LOH Region Score + 0.67 * TAI Region Score + 0.12 * LST Region Score
  • the present invention provides a method of predicting the status of BRCA1 and BRCA2 genes in a sample. Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to assess ( e.g. , detect) BRCA1 and/or BRCA2 deficiency in the sample.
  • this invention provides a method of predicting a cancer patient's response to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor.
  • Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to predict the likelihood that the cancer patient will respond to the cancer treatment regimen.
  • the patients are treatment naive patients.
  • this invention provides a method of treating cancer. Such method is analogous to the methods described above and differs in that a particular treatment regimen is administered (recommended, prescribed, etc.) based at least in part on the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these.
  • this invention features the use of one or more drugs selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors, in the manufacture of a medicament useful for treating a cancer in a patient identified as having (or as having had) a cancer cell determined to have HRD (e.g., an HRD signature) as described herein.
  • this document features a method for assessing a sample for the presence of a mutation within a gene from an HDR pathway.
  • Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to detect (or not) the presence of a mutation within a gene from an HDR pathway.
  • the invention provides a method for assessing a patient.
  • the method comprises, or consists essentially of, (a) determining whether the patient has (or had) cancer cells with more than a reference number of CA Regions (or, e.g., a CA Region Score exceeding a reference CA Region Score); and (b)(1) diagnosing the patient as having cancer cells with HRD if it is determined that the patient has (or had) cancer cells with more than a reference number of CA Regions (or, e.g., a CA Region Score exceeding a reference CA Region Score); or (b)(2) diagnosing the patient as not having cancer cells with HRD if it is determined that the patient does not have (or has not had) cancer cells with more than a reference number of CA Regions (or, e.g., the patient does not have (or has not had) cancer cells with a CA Region Score exceeding a reference CA Region Score).
  • this invention features the use of a plurality of oligonucleotides capable of hybridizing to a plurality of polymorphic regions of human genomic DNA, in the manufacture of a diagnostic kit useful for determining the total number or combined length of CA Regions in at least a chromosome pair (or DNA derived therefrom) in a sample obtained from a cancer patient, and for detecting (a) HRD or likelihood of HRD (e.g., an HRD signature) in the sample, (b) deficiency (or likelihood of deficiency) in a BRCA1 or BRCA2 gene in the sample, or (c) an increased likelihood that the cancer patient will respond to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, or a PARP inhibitor.
  • a DNA damaging agent e.g., an an HRD signature
  • this invention features a system for detecting HRD (e.g., an HRD signature) in a sample.
  • the system comprises, or consists essentially of, (a) a sample analyzer configured to produce a plurality of signals about genomic DNA of at least one pair of human chromosomes (or DNA derived therefrom) in the sample, and (b) a computer sub-system programmed to calculate, based on the plurality of signals, the number or combined length of CA Regions in the at least one pair of human chromosomes.
  • the computer sub-system can be programmed to compare the number or combined length of CA Regions to a reference number to detect (a) HRD or likelihood of HRD (e.g., an HRD signature) in the sample, (b) deficiency (or likelihood of deficiency) in a BRCA1 or BRCA2 gene in the sample, or (c) an increased likelihood that the cancer patient will respond to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, or a PARP inhibitor.
  • the system can comprise an output module configured to display (a), (b), or (c).
  • the system can comprise an output module configured to display a recommendation for the use of the cancer treatment regimen.
  • the invention provides a computer program product embodied in a computer readable medium that, when executing on a computer, provides instructions for detecting the presence or absence of any CA Region along one or more of human chromosomes other than the human X and Y sex chromosomes (the CA Regions optionally being Indicator CA Regions); and determining the total number or combined length of the CA Regions in the one or more chromosome pairs.
  • the computer program product can include other instructions.
  • the present invention provides a diagnostic kit.
  • the kit comprises, or consists essentially of, at least 500 oligonucleotides capable of hybridizing to a plurality of polymorphic regions of human genomic DNA (or DNA derived therefrom); and a computer program product provided herein.
  • the computer program product can be embodied in a computer readable medium that, when executing on a computer, provides instructions for detecting the presence or absence of any CA Region along one or more of human chromosomes other than the human X and Y sex chromosomes (the CA Regions optionally being Indicator CA Regions); and determining the total number or combined length of the CA Regions in the one or more chromosome pairs.
  • the computer program product can include other instructions.
  • the CA Regions can be determined in at least two, five, ten, or 21 pairs of human chromosomes.
  • the cancer cell can be an ovarian, breast, lung or esophageal cancer cell.
  • the reference can be 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 20 or greater.
  • the at least one pair of human chromosomes can exclude human chromosome 17.
  • the DNA damaging agent can be cisplatin, carboplatin, oxalaplatin, or picoplatin
  • the anthracycline can be epirubincin or doxorubicin
  • the topoisomerase I inhibitor can be campothecin, topotecan, or irinotecan
  • the PARP inhibitor can be iniparib, olaparib or velapirib.
  • the patient can be a treatment naive patient.
  • one aspect of this invention features a method for assessing HRD in a cancer cell or DNA (e.g. , genomic DNA) derived therefrom.
  • the method comprises, or consists essentially of, (a) detecting, in a sample or DNA derived therefrom, CA Regions in at least one pair of human chromosomes or DNA derived therefrom; and (b) determining the number, size ( e.g., length), and/or character of said CA Regions.
  • chromosomal aberration means a somatic change in a cell's chromosomal DNA that falls into at least one of three overlapping categories: LOH, TAI, or LST.
  • Polymorphic loci within the human genome e.g. , single nucleotide polymorphisms (SNPs)
  • SNPs single nucleotide polymorphisms
  • LOH loss of heterozygosity
  • a locus of one chromosome can be deleted in a somatic cell.
  • the locus that remains present on the other chromosome is an LOH locus as there is only one copy (instead of two copies) of that locus present within the genome of the affected cells. This type of LOH event results in a copy number reduction.
  • a locus of one chromosome e.g. , one non-sex chromosome for males
  • a locus of one chromosome in a somatic cell can be replaced with a copy of that locus from the other chromosome, thereby eliminating any heterozygosity that may have been present within the replaced locus.
  • LOH locus that remains present on each chromosome is an LOH locus and can be referred to as a copy neutral LOH locus.
  • LOH and its use in determining HRD is described in detail in International Application no. PCT/US2011/040953 (published as WO/2011/160063 ), the entire contents of which are incorporated herein by reference.
  • allelic imbalance occurs when the relative copy number (i.e., copy proportion) at a particular locus in somatic cells differs from the germline. For example, if the germline has one copy of allele A and one copy of allele B at a particular locus, and a somatic cell has two copies of A and one copy of B, there is allelic imbalance at the locus because the copy proportion of the somatic cell (2:1) differs from the germline (1:1). LOH is an example of allelic imbalance since the somatic cell has a copy proportion (1:0 or 2:0) that differs from the germline (1:1).
  • allelic imbalance encompasses more types of chromosomal aberration, e.g. , 2:1 germline going to 1:1 somatic; 1:0 germline going to 1:1 somatic; 1:1 germline going to 2:1 somatic, etc.
  • Analysis of regions of allelic imbalance encompassing the telomeres of chromosomes is particularly useful in the invention.
  • a "telomeric allelic imbalance region" or "TAI Region” is defined as a region with allelic imbalance that (a) extends to one of the subtelomeres and (b) does not cross the centromere. TAI and its use in determining HRD is described in detail in International Application no. PCT/US2011/048427 (published as WO/2012/027224 ), the entire contents of which are incorporated herein by reference.
  • LST large scale transition
  • LST refers to any somatic copy number transition (i.e., breakpoint) along the length of a chromosome where it is between two regions of at least some minimum length (e.g., at least 3, 4, 5, 6, 7, 8 9, 10, 11 12, 13, 14, 15, 16, 17, 18, 19 or 20 or more megabases) after filtering out regions shorter than some maximum length (e.g., 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4 or more megabases).
  • LST Region is genomic region with stable copy number across at least some minimum length (e.g., at least 3, 4, 5, 6, 7, 8, 9, 10, 11 12, 13, 14, 15, 16, 17, 18, 19 or 20 megabases) bounded by breakpoints (i.e., transitions) where the copy number changes for another region also at least this minimum length.
  • the somatic cell has a region of at least 10 megabases with copy number of 1:1 bounded on one side by a breakpoint transition to a region of, e.g., at least 10 megabases with copy number 2:2, and bounded on the other side by a breakpoint transition to a region of, e.g., at least 10 megabases with copy number 1:2, then this is two LSTs. Notice that this is broader than allelic imbalance because such a copy number change would not be considered allelic imbalance (because the copy proportions 1:1 and 2:2 are the same, i.e., there has been no change in copy proportion).
  • LST score may be used for "near-diploid" and "near-tetraploid" tumors to separate BRCA1/2 intact and deficient samples. LST score sometimes increases with ploidy both within intact and deficient samples.
  • CA Regions can be any length (e.g., from a length less than about 1.5 Mb up to a length equal to the entire length of the chromosome).
  • An abundance of large CA Regions (“Indicator CA Regions") indicate a deficiency in the homology-dependent repair (HDR) mechanism of a cell.
  • HDR homology-dependent repair
  • an "LOH Region” means at least some minimum number of consecutive loci exhibiting LOH or some minimum stretch of genomic DNA having consecutive loci exhibiting LOH.
  • a "TAI Region,” on the other hand, means at least some minimum number of consecutive loci exhibiting allelic imbalance extending from the telomere into the rest of the chromosome (or some minimum stretch of genomic DNA extending from the telomere into the rest of the chromosome having consecutive loci exhibiting allelic imbalance).
  • LST is already defined in terms of a region of genomic DNA of at least some minimum size, so "LST” and “LST Region” are used interchangeably in this document to refer to a minimum number of consecutive loci (or some minimum stretch of genomic DNA) having the same copy number bounded by a breakpoint or transition from that copy number to a different one.
  • a CA Region (whether an LOH Region, TAI region, or LST Region) is an Indicator CA Region (whether an Indicator LOH Region, Indicator TAI region, or Indicator LST Region) if it is at least 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 45, 50, 60, 70, 80, 90, or 100 megabases or more in length.
  • Indicator LOH Regions are LOH Regions that are longer than about 1.5, 5, 12, 13, 14, 15, 16, 17 or more (preferably 14, 15, 16 or more, more preferably 15 or more) megabases but shorter than the entire length of the respective chromosome within which the LOH Region is located.
  • Indicator TAI Regions are TAI Regions with allelic imbalance that (a) extend to one of the subtelomeres, (b) do not cross the centromere and (c) are longer than 1.5, 5, 12, 13, 14, 15, 16, 17 or more (preferably 10, 11, 12 or more, more preferably 11 or more) megabases.
  • the total combined length of such Indicator TAI Regions may be determined. Because the concept of LST already involves regions of some minimum size (such minimum size being determined based on its ability to differentiate HRD from HDR intact samples), Indicator LST Regions as used herein are the same as LST Regions.
  • an LST Region Score can be either derived from the number of regions showing LST as described above or the number of LST breakpoints.
  • the minimum length of the region of stable copy number bounding the LST breakpoint is at least 3, 4, 5, 6, 7, 8, 9, 10, 11 12, 13, 14, 15, 16, 17, 18, 19 or 20 megabases (preferably 8, 9, 10, 11 or more megabases, more preferably 10 megabases) and the maximum region remaining unfiltered is less than 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4 or fewer megabases (preferably 2, 2.5, 3, 3.5, or 4 or fewer megabases, more preferably fewer than 3 megabases).
  • a sample has an "HRD signature" if such sample has a number of Indicator CA Regions (as described herein) or a CA Region Score (as described herein) exceeding a reference as described herein, wherein a number or score exceeding such reference indicates homologous recombination deficiency.
  • the invention generally involves detecting and quantifying Indicator CA Regions in a sample to determine whether cells in the sample (or cells from which DNA in the sample are derived) have HRD. Often this comprises comparing the number of Indicator CA Regions (or a test value or score derived or calculated therefrom and corresponding to such number) to a reference or index number (or score).
  • the various aspects of the present invention comprise using a combined analysis of two or more types of CA Regions (including two or more types of Indicator CA Regions) to assess ( e.g., detect, diagnose) HRD in a sample.
  • the invention provides a method of assessing ( e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining the total number (or combined length) of Indicator LOH Regions in the sample; (2) determining the total number (or combined length) of Indicator TAI Regions in the sample; and (3) determining the presence or absence of ( e.g., detecting, diagnosing) HRD in the sample based at least in part on the determinations made in (1) and (2).
  • the invention provides a method of assessing (e.g., detecting, diagnosing) HRD in a sample comprising (1) determining the total number (or combined length) of Indicator LOH Regions in the sample; (2) determining the total number (or combined length) of Indicator LST Regions in the sample; and (3) determining the presence or absence of ( e.g., detecting, diagnosing) HRD in the sample based at least in part on the determinations made in (1) and (2).
  • the invention provides a method of assessing (e.g., detecting, diagnosing) HRD in a sample comprising (1) determining the total number (or combined length) of Indicator TAI Regions in the sample; (2) determining the total number (or combined length) of Indicator LST Regions in the sample; and (3) determining the presence or absence of ( e.g., detecting, diagnosing) HRD in the sample based at least in part on the determinations made in (1) and (2).
  • the invention provides a method of assessing (e.g., detecting, diagnosing) HRD in a sample comprising (1) determining the total number (or combined length) of Indicator LOH Regions in the sample; (2) determining the total number of Indicator TAI Regions in the sample; (3) determining the total number (or combined length) of Indicator LST Regions in the sample; and (4) determining the presence or absence of ( e.g., detecting, diagnosing) HRD in the sample based at least in part on the determinations made in (1), (2) and (3).
  • the various aspects of the present invention comprise using a combined analysis of the averages of three different CA Regions to assess ( e.g. , detect, diagnose) HRD in a sample.
  • the invention provides a method of assessing ( e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of TAI Regions of a certain size or character ( e.g. , "Indicator TAI Regions", as defined herein) in the sample; (3) determining the total number of LST Regions of a certain size or character (e.g.
  • CA Region Score means a test value or score derived or calculated from ( e.g., representing or corresponding to) Indicator CA Regions detected in a sample ( e.g., a score or test value derived or calculated from the number of Indicator CA Regions detected in a sample).
  • LOH Region Score is a subset of CA Region Scores and means a test value or score derived or calculated from ( e.g. , representing or corresponding to) Indicator LOH Regions detected in a sample ( e.g. , a score or test value derived or calculated from the number of Indicator LOH Regions detected in a sample), and so on for TAI Region Score and LST Region Score.
  • Such a score may in some embodiments be simply the number of Indicator CA Regions detected in a sample. In some embodiments the score is more complicated, factoring in the lengths of each Indicator CA Region or a subset of Indicator CA Regions detected.
  • the invention will generally involve combining the analysis of two or more types of CA Region Scores (which may include the number of such regions).
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on either the LOH Region Score exceeding a reference or the TAI Region Score exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on both the LOH Region Score not exceeding a reference and the TAI Region Score not exceeding a reference.
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on either the LOH Region exceeding a reference or the LST Region Score exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on both the LOH Region Score not exceeding a reference and the LST Region Score not exceeding a reference.
  • the invention provides a method of assessing (e.g.
  • detecting, diagnosing) HRD in a sample comprising (1) determining a TAI Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on either the TAI Region Score exceeding a reference or the LST Region Score exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on both the TAI Region Score not exceeding a reference and the LST Region Score not exceeding a reference.
  • the invention provides a method of assessing (e.g.
  • detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample; (3) determining an LST Region Score for the sample; and (4)(a) detecting (or diagnosing) HRD in the sample based at least in part on either the LOH Region Score exceeding reference, the TAI Region Score exceeding a reference or the LST Region Score exceeding a reference; or optionally (4)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on the LOH Region Score not exceeding a reference, the TAI Region Score not exceeding a reference and the LST Region Score not exceeding a reference.
  • the CA Region Score is a combination of scores derived or calculated from ( e.g. , representing or corresponding to) two or more of (1) the detected LOH Regions ("LOH Region Score", as defined herein), (2) the detected TAI Regions ("TAI Region Score", as defined herein), and/or (3) the detected LST Regions ("LST Region Score", as defined herein).
  • CA Region Score 0.32 * LOH Region Score + 0.68 * TAI Region Score
  • CA Region Score 0.34 * LOH Region Score + 0.66 * TAI Region Score
  • CA Region Score A * LOH Region Score + B * LST Region Score
  • CA Region Score A * TAI Region Score + B * LST Region Score
  • CA Region Score A * LOH Region Score + B * TAI Region Score + C * LST Region Score
  • CA Region Score 0.21 * LOH Region Score + 0.67 * TAI Region Score + 0.12 * LST Region Score
  • CA Region Score 0.24 * LOH Region Score + 0.65 * TAI Region Score + 0.11 * LST Region Score
  • CA Region Score 0.11 * LOH Region Score + 0.25 * TAI Region Score + 0.12 * LST Region Score
  • CA Region Score A * LOH Region Score + B * TAI Region Score 2
  • CA Region Score A * LOH Region Score + C * LST Region Score 2
  • CA Region Score B * TAI Region Score + C * LST Region Score 2
  • one or more of these coefficients is 1 and in some embodiments all three coefficients (i.e., A, B, and C) are 1.
  • a formula may not have all of the specified coefficients (and thus not incorporate the corresponding variable(s)).
  • the embodiment mentioned immediately previously may be applied to formula (2) where A in formula (2) is 0.95 and B in formula (2) is 0.61. C and D would not be applicable as these coefficients and their corresponding variables are not found in formula (2) (though the clinical variables are incorporated into the clinical score found in formula (2)).
  • A is between 0.9 and 1, 0.9 and 0.99, 0.9 and 0.95, 0.85 and 0.95, 0.86 and 0.94, 0.87 and 0.93, 0.88 and 0.92, 0.89 and 0.91, 0.85 and 0.9, 0.8 and 0.95, 0.8 and 0.9, 0.8 and 0.85, 0.75 and 0.99, 0.75 and 0.95, 0.75 and 0.9, 0.75 and 0.85, or between 0.75 and 0.8.
  • B is between 0.40 and 1, 0.45 and 0.99, 0.45 and 0.95, 0.55 and 0.8, 0.55 and 0.7, 0.55 and 0.65, 0.59 and 0.63, or between 0.6 and 0.62.
  • C is, where applicable, between 0.9 and 1, 0.9 and 0.99, 0.9 and 0.95, 0.85 and 0.95, 0.86 and 0.94, 0.87 and 0.93, 0.88 and 0.92, 0.89 and 0.91, 0.85 and 0.9, 0.8 and 0.95, 0.8 and 0.9, 0.8 and 0.85, 0.75 and 0.99, 0.75 and 0.95, 0.75 and 0.9, 0.75 and 0.85, or between 0.75 and 0.8.
  • D is, where applicable, between 0.9 and 1, 0.9 and 0.99, 0.9 and 0.95, 0.85 and 0.95, 0.86 and 0.94, 0.87 and 0.93, 0.88 and 0.92, 0.89 and 0.91, 0.85 and 0.9, 0.8 and 0.95, 0.8 and 0.9, 0.8 and 0.85, 0.75 and 0.99, 0.75 and 0.95, 0.75 and 0.9, 0.75 and 0.85, or between 0.75 and 0.8.
  • A is between 0.1 and 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 20; or between 0.2 and 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 20; or between 0.3 and 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 20; or between 0.4 and 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, or 20; or between 0.4 and 0.5, 0.6, 0.7, 0.8, 0.9, 1, 1.5, 2, 2.5, 3, 3.5, 4, 4.5, 5, 6, 7, 8, 9, 10, 11, 12, 13,
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on a combination of the LOH Region Score and the TAI Region Score ( e.g., a Combined CA Region Score) exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on a combination of the LOH Region Score and the TAI Region Score (e.g., a Combined CA Region Score) not exceeding a reference.
  • a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on a combination of the LOH Region Score and the LST Region Score ( e.g., a Combined CA Region Score) exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on a combination of the LOH Region Score and the LST Region Score (e.g., a Combined CA Region Score) not exceeding a reference.
  • a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining a TAI Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in part on a combination of the TAI Region Score and the LST Region Score ( e.g., a Combined CA Region Score) exceeding a reference; or optionally (3)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on a combination of the TAI Region Score and the LST Region Score (e.g., a Combined CA Region Score) not exceeding a reference.
  • a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining a TAI Region Score for the sample; (2) determining an LST Region Score for the sample; and (3)(a) detecting (or diagnosing) HRD in the sample based at least in
  • the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample; (3) determining an LST Region Score for the sample; and (4)(a) detecting (or diagnosing) HRD in the sample based at least in part on a combination of the LOH Region Score, the TAI Region Score and the LST Region Score ( e.g., a Combined CA Region Score) exceeding a reference; or optionally (4)(b) detecting (or diagnosing) an absence of HRD in the sample based at least in part on the LOH Region Score, the TAI Region Score and the LST Region Score ( e.g., a Combined CA Region Score) not exceeding a reference.
  • a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining an LOH Region Score for the sample; (2) determining a TAI Region Score for the sample
  • Another aspect of the invention provides a method of assessing (e.g. , detecting, diagnosing) HRD in a sample comprising (1) determining the total number of LOH Regions of a certain size or character (e.g. , "Indicator LOH Regions", as defined herein) in the sample; (2) determining the total number of TAI Regions of a certain size or character (e.g. , "Indicator TAI Regions", as defined herein) in the sample; (3) determining the total number of LST Regions of a certain size or character ( e.g., "Indicator LST Regions", as defined herein) in the sample; (4) calculating the average ( e.g. , arithmetic mean) of the determinations made in (1), (2), and (3); and (5) assessing HRD in the sample based at least in part on the calculated average ( e.g. , arithmetic mean) made in (4).
  • the reference (or index) discussed above for the CA Region Score may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, 19, 20 or greater, preferably 5, preferably 8, more preferably 9 or 10, most preferably 10.
  • the reference for the total e.g.
  • Indicator CA Regions may be about 75, 90, 105, 120, 130, 135, 150, 175, 200, 225, 250, 275, 300, 325 350, 375, 400, 425, 450, 475, 500 megabases or greater, preferably about 75 megabases or greater, preferably about 90 or 105 megabases or greater, more preferably about 120 or 130 megabases or greater, and more preferably about 135 megabases or greater, and most preferably about 150 megabases or greater.
  • the reference discussed above for the Combined CA Region Score (e.g., the combined number of Indicator LOH Regions, Indicator, TAI Regions and/or Indicator LST Regions) may be 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 18, 19, 20 or greater, preferably 5, preferably 8, more preferably 9 or 10, most preferably 10.
  • the reference for the total (e.g.
  • Indicator LOH Regions, Indicator TAI Regions and/or Indicator LST Regions may be about 75, 90, 105, 120, 130, 135, 150, 175, 200, 225, 250, 275, 300, 325 350, 375, 400, 425, 450, 475, 500 megabases or greater, preferably about 75 megabases or greater, preferably about 90 or 105 megabases or greater, more preferably about 120 or 130 megabases or greater, and more preferably about 135 megabases or greater, and most preferably about 150 megabases or greater
  • the number of Indicator CA Regions (or the combined length, a CA Region Score or a Combined CA Region Score) in a sample is considered "greater” than a reference if it is at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10-fold greater than the reference while in some embodiments, it is considered “greater” if it is at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 standard deviations greater than the reference.
  • the number of Indicator CA Regions (or the combined length, a CA Region Score or a Combined CA Region Score) in a sample is considered “not greater” than a reference if it is not more than 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10-fold greater than the reference while in some embodiments, it is considered “not greater” if it is not more than 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 standard deviations greater than the reference.
  • the reference number is derived from a relevant reference population.
  • Such reference populations may include patients (a) with the same cancer as the patient being tested, (b) with the same cancer sub-type, (c) with cancer having similar genetic or other clinical or molecular features, (d) who responded to a particular treatment, (e) who did not respond to a particular treatment, (f) who are apparently healthy ( e.g., do not have any cancer or at least do not have the tested patient's cancer), etc.
  • the reference number may be (a) representative of the number (or length, value or score) found in the reference population as a whole, (b) an average (mean, median, etc.) of the number (or length, value or score) found in the reference population as a whole or a particular sub-population, (c) representative of the number (or length, value or score) ( e.g., an average such as mean or median) found in terciles, quartiles, quintiles, etc. of the reference population as ranked by (i) their respective number (or length, value or score) or (ii) the clinical feature they were found to have ( e.g. , strength of response, prognosis (including time to cancer-specific death), etc.).
  • the reference or index that, if exceeded by the test value or score from the sample, indicates HRD is the same as the reference that, if not exceeded by the test value or score from the sample, indicates the absence of HRD (or functional HDR). In some embodiments they are different.
  • the present invention provides a method of predicting the status of BRCA1 and BRCA2 genes in a sample.
  • Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to assess ( e.g. , detect) BRCA1 and/or BRCA2 deficiency in the sample.
  • this invention provides a method of predicting a cancer patient's response to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor.
  • a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor.
  • Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to predict the likelihood that the cancer patient will respond to the cancer treatment regimen.
  • the patients are treatment naive patients.
  • this invention provides a method of treating cancer. Such method is analogous to the methods described above and differs in that a particular treatment regimen is administered (recommended, prescribed, etc.) based at least in part on the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these.
  • this invention features the use of one or more drugs selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors, in the manufacture of a medicament useful for treating a cancer in a patient identified as having (or as having had) a cancer cell determined to have HRD (e.g., an HRD signature) as described herein.
  • drugs selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors
  • this document features a method for assessing a sample for the presence of a mutation within a gene from an HDR pathway.
  • Such method is analogous to the methods described above and differs in that the determination of CA Regions, LOH Regions, TAI Regions, LST Regions, or scores incorporating these are used to detect (or not) the presence of a mutation within a gene from an HDR pathway.
  • this document features a method for assessing cancer cells of a patient for the presence of an HRD signature.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient, and (b) identifying the patient as having cancer cells with the HRD signature.
  • this document features a method for assessing cancer cells of a patient for the presence of an HDR deficient status.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient, and (b) identifying the patient as having cancer cells with the HDR deficient status.
  • this document features a method for assessing cancer cells of a patient for the presence of a genetic mutation within a gene from an HDR pathway.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient, and (b) identifying the patient as having cancer cells with the genetic mutation.
  • this document features a method for determining if a patient is likely to respond to a cancer treatment regimen comprising administering radiation or a drug selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient, and (b) identifying the patient as being likely to respond to the cancer treatment regimen.
  • this document features a method for assessing a patient.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having an HRD signature, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HRD signature, and (b) diagnosing the patient as having cancer cells with the HRD signature.
  • this document features a method for assessing a patient.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having an HDR deficiency status, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HDR deficiency status, and (b) diagnosing the patient as having cancer cells with the HDR deficient status.
  • this document features a method for assessing a patient.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having a genetic mutation within a gene from an HDR pathway, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the genetic mutation, and (b) diagnosing the patient as having cancer cells with the genetic mutation.
  • this document features a method for assessing a patient for a likelihood to respond to a cancer treatment regimen comprising administering radiation or a drug selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having an HRD signature, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HRD signature, and (b) diagnosing, based at least in part on the presence of the HRD signature, the patient as being likely to respond to the cancer treatment regimen.
  • this document features a method for performing a diagnostic analysis of a cancer cell of a patient.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of the cancer cell, and (b) identifying or classifying the patient as having cancer cells with an HRD signature.
  • this document features a method for performing a diagnostic analysis of a cancer cell of a patient.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of the cancer cell, and (b) identifying or classifying the patient as having cancer cells with a HDR deficient status.
  • this document features a method for performing a diagnostic analysis of a cancer cell of a patient.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of the cancer cell that are longer, and (b) identifying or classifying the patient as having cancer cells with a genetic mutation within a gene from an HDR pathway.
  • this document features a method for performing a diagnostic analysis of a cancer cell of a patient to determine if the cancer patient is likely to respond to a cancer treatment regimen comprising administering radiation or a drug selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors.
  • the method comprises, or consists essentially of, (a) detecting the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of the cancer cell, and (b) identifying or classifying the patient as being likely to respond to the cancer treatment regimen.
  • this document features a method for diagnosing a patient as having cancer cells having an HRD signature.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having the HRD signature, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HRD signature, and (b) diagnosing the patient as having cancer cells with the HRD signature.
  • this document features a method for diagnosing a patient as having cancer cells with an HDR deficient status.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having the HDR deficiency status, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HDR deficiency status, and (b) diagnosing the patient as having cancer cells with the HDR deficient status.
  • this document features a method for diagnosing a patient as having cancer cells with a genetic mutation within a gene from an HDR pathway.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having the genetic mutation, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the genetic mutation, and (b) diagnosing the patient as having cancer cells with the genetic mutation.
  • this document features a method for diagnosing a patient as being a candidate for a cancer treatment regimen comprising administering radiation or a drug selected from the group consisting of DNA damaging agents, anthracyclines, topoisomerase I inhibitors, and PARP inhibitors.
  • the method comprises, or consists essentially of, (a) determining that the patient comprises cancer cells having an HRD signature, wherein the presence of more than a reference number of Indicator CA Regions in at least one pair of human chromosomes of a cancer cell of the cancer patient indicates that the cancer cells have the HRD signature, and (b) diagnosing, based at least in part on the presence of the HRD signature, the patient as being likely to respond to the cancer treatment regimen.
  • the invention provides a method for assessing a patient.
  • the method comprises, or consists essentially of, (a) determining whether the patient has (or had) cancer cells with more than a reference number of Indicator CA Regions (or, e.g., a CA Region Score exceeding a reference CA Region Score); and (b)(1) diagnosing the patient as having cancer cells with HRD if it is determined that the patient has (or had) cancer cells with more than a reference number of CA Regions (or, e.g., a CA Region Score exceeding a reference CA Region Score); or (b)(2) diagnosing the patient as not having cancer cells with HRD if it is determined that the patient does not have (or has not had) cancer cells with more than a reference number of CA Regions (or, e.g., the patient does not have (or has not had) cancer cells with a CA Region Score exceeding a reference CA Region Score).
  • this invention features the use of a plurality of oligonucleotides capable of hybridizing to a plurality of polymorphic regions of human genomic DNA, in the manufacture of a diagnostic kit useful for determining the total number or combined length of CA Regions in at least a chromosome pair (or DNA derived therefrom) in a sample obtained from a cancer patient, and for detecting (a) HRD or likelihood of HRD (e.g., an HRD signature) in the sample, (b) deficiency (or likelihood of deficiency) in a BRCA1 or BRCA2 gene in the sample, or (c) an increased likelihood that the cancer patient will respond to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, or a PARP inhibitor.
  • a DNA damaging agent e.g., an an HRD signature
  • this invention features a system for detecting HRD (e.g., an HRD signature) in a sample.
  • the system comprises, or consists essentially of, (a) a sample analyzer configured to produce a plurality of signals about genomic DNA of at least one pair of human chromosomes (or DNA derived therefrom) in the sample, and (b) a computer sub-system programmed to calculate, based on the plurality of signals, the number or combined length of CA Regions in the at least one pair of human chromosomes.
  • the computer sub-system can be programmed to compare the number or combined length of CA Regions to a reference number to detect (a) HRD or likelihood of HRD (e.g., an HRD signature) in the sample, (b) deficiency (or likelihood of deficiency) in a BRCA1 or BRCA2 gene in the sample, or (c) an increased likelihood that the cancer patient will respond to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, or a PARP inhibitor.
  • the system can comprise an output module configured to display (a), (b), or (c).
  • the system can comprise an output module configured to display a recommendation for the use of the cancer treatment regimen.
  • the invention provides a computer program product embodied in a computer readable medium that, when executing on a computer, provides instructions for detecting the presence or absence of any CA Region along one or more of human chromosomes other than the human X and Y sex chromosomes (the CA Regions optionally being Indicator CA Regions); and determining the total number or combined length of the CA Regions in the one or more chromosome pairs.
  • the computer program product can include other instructions.
  • the present invention provides a diagnostic kit.
  • the kit comprises, or consists essentially of, at least 500 oligonucleotides capable of hybridizing to a plurality of polymorphic regions of human genomic DNA (or DNA derived therefrom); and a computer program product provided herein.
  • the computer program product can be embodied in a computer readable medium that, when executing on a computer, provides instructions for detecting the presence or absence of any CA Region along one or more of human chromosomes other than the human X and Y sex chromosomes (the CA Regions optionally being Indicator CA Regions); and determining the total number or combined length of the CA Regions in the one or more chromosome pairs.
  • the computer program product can include other instructions.
  • the CA Regions can be determined in at least two, five, ten, or 21 pairs of human chromosomes.
  • the cancer cell can be an ovarian, breast, lung or esophageal cancer cell.
  • the reference can be 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 20 or greater.
  • the at least one pair of human chromosomes can exclude human chromosome 17.
  • the DNA damaging agent can be cisplatin, carboplatin, oxalaplatin, or picoplatin
  • the anthracycline can be epirubincin or doxorubicin
  • the topoisomerase I inhibitor can be campothecin, topotecan, or irinotecan
  • the PARP inhibitor can be iniparib, olaparib or velapirib.
  • the patient can be a treatment naive patient.
  • a sample e.g. , cancer cell sample or a sample containing DNA derived from one or more cancer cells
  • HRD signature or alternatively called “HDR-deficiency signature”
  • the genome of the cells being assessed contains (a) any of an LOH Region Score, a TAI Region Score or an LST Region Score exceeding a reference or (b) a Combined CA Region Score exceeding a reference.
  • a sample e.g.
  • cancer cell sample or a sample containing DNA derived from one or more cancer cells can be identified as lacking an "HRD signature" (or alternatively called “HDR-deficiency signature") if the genome of the cells being assessed contains (a) an LOH Region Score, a TAI Region Score and an LST Region Score each not exceeding a reference or (b) a Combined CA Region Score not exceeding a reference.
  • HRD signature or alternatively called "HDR-deficiency signature
  • Cells identified as having an HRD signature can be classified as having an increased likelihood of having an HDR deficiency and/or as having an increased likelihood of having a deficient status in one or more genes in the HDR pathway.
  • cancer cells identified as having an HRD signature can be classified as having an increased likelihood of having an HDR deficient status.
  • cancer cells identified as having an HRD signature can be classified as having an increased likelihood of having a deficient status for one or more genes in the HDR pathway.
  • deficient status for a gene means the sequence, structure, expression and/or activity of the gene or its product is/are deficient as compared to normal.
  • deficient status for a pathway means at least one gene in that pathway (e.g. , BRCA1) is deficient.
  • highly deleterious mutations include frameshift mutations, stop codon mutations, and mutations that lead to altered RNA splicing.
  • Deficient status in a gene in the HDR pathway may result in deficiency or reduced activity in homology directed repair in the cancer cells. Examples of genes in the HDR pathway include, without limitation, the genes listed in Table 1. Table 1.
  • identifying CA loci can include, first, determining the genotype of a sample at various genomic loci (e.g., SNP loci, individual bases in large-scale sequencing) and, second, determining whether the loci exhibit any of LOH, TAI or LST.
  • Any appropriate technique can be used to determine genotypes at loci of interest within the genome of a cell. For example, single nucleotide polymorphism (SNP) arrays (e.g., human genome-wide SNP arrays), targeted sequencing of loci of interest (e.g., sequencing SNP loci and their surrounding sequences), and even large-scale sequencing (e.g.
  • SNP single nucleotide polymorphism
  • loci can be homozygous or heterozygous.
  • an analysis of the homozygous or heterozygous nature of loci over a length of a chromosome can be performed to determine the length of CA Regions. For example, a stretch of SNP locations that are spaced apart ( e.g. , spaced about 25 kb to about 100 kb apart) along a chromosome can be evaluated using SNP array results to determine not only the presence of a region of homozygosity (e.g. , LOH) along a chromosome but also the length of that region.
  • a region of homozygosity e.g. , LOH
  • Results from a SNP array can be used to generate a graph that plots allele dosages along a chromosome.
  • Figures 1 and 2 show the difference between fresh frozen and FFPE samples and between SNP microarray and SNP sequencing analyses.
  • Numerous variations on nucleic acid arrays useful in the invention are known in the art. These include the arrays used in the various examples below ( e.g. , Affymetrix 500K GeneChip array in Example 3; Affymetrix OncoScanTM FFPE Express 2.0 Services (Formerly MIP CN Services) in Example 4).
  • determining whether chromosomal imbalance or large scale transitions includes determining whether these are somatic or germline aberrations.
  • the genotype for a plurality of loci can be determined in both a germline (e.g., blood) sample and a somatic (e.g., tumor) sample.
  • the genotypes for each sample can be compared (typically computationally) to determine where the genome of the germline cell was heterozygous and the genome of the somatic cell is homozygous.
  • loci are LOH loci and regions of such loci are LOH Regions.
  • Computational techniques can also be used to determine whether an aberration is germline or somatic. Such techniques are particularly useful when a germline sample is not available for analysis and comparison. For example, algorithms such as those described elsewhere can be used to detect LOH regions using information from SNP arrays ( Nannya et al., Cancer Res. (2005) 65:6071-6079 (2005 )). Typically these algorithms do not explicitly take into account contamination of tumor samples with benign tissue. Cf International Application No. PCT/US2011/026098 to Abkevich et al. ; Goransson et al., PLoS One (2009) 4(6):e6057 . This contamination is often high enough to make the detection of LOH regions challenging. Improved analytical methods according to the present invention for identifying LOH, TAI and LST, even in spite of contamination, include those embodied in computer software products as described below.
  • the first possibility is that cancer cells have LOH with deletion of allele B in a sample with 50% contamination with normal cells.
  • the second possibility is that there is no LOH but allele A is duplicated in a sample with no contamination with normal cells.
  • An algorithm can be implemented as a computer program as described herein to reconstruct LOH regions based on genotype (e.g., SNP genotype) data.
  • One point of the algorithm is to first reconstruct allele specific copy numbers (ASCN) at each locus (e.g., SNP).
  • ASCNs are the numbers of copies of both paternal and maternal alleles.
  • An LOH region is then determined as a stretch of SNPs with one of the ASCNs (paternal or maternal) being zero.
  • the algorithm can be based on maximizing a likelihood function and can be conceptually akin to a previously described algorithm designed to reconstruct total copy number (rather than ASCN) at each locus ( e.g., SNP). See International Application No. PCT/US2011/026098 to Abkevich et al.
  • the likelihood function can be maximized over ASCN of all loci, level of contamination with benign tissue, total copy number averaged over the whole genome, and sample specific noise level.
  • the input data for the algorithm can include or consist of (1) sample-specific normalized signal intensities for both allele of each locus and (2) assay-specific (specific for different SNP arrays and for sequence based approach) set of parameters defined based on analysis of large number of samples with known ASCN profiles.
  • nucleic acid sequencing techniques can be used to genotype loci.
  • genomic DNA from a cell sample e.g. , a cancer cell sample
  • Any appropriate method can be used to extract and fragment genomic nucleic acid including, without limitation, commercial kits such as QIAampTM DNA Mini Kit (QiagenTM), MagNATM Pure DNA Isolation Kit (Roche Applied ScienceTM) and GenEluteTM Mammalian Genomic DNA Miniprep Kit (Sigma-AldrichTM).
  • QIAampTM DNA Mini Kit QiagenTM
  • MagNATM Pure DNA Isolation Kit Roche Applied ScienceTM
  • GenEluteTM Mammalian Genomic DNA Miniprep Kit Sigma-AldrichTM
  • either targeted or untargeted sequencing can be done to determine the sample's genotypes at loci.
  • whole genome, whole transcriptome, or whole exome sequencing can be done to determine genotypes at millions or even billions of base pairs ( i.e., base pairs can be "loci" to be evaluated).
  • genomic DNA can be enriched for those fragments containing a locus (e.g., SNP location) to be analyzed using kits designed for this purpose (e.g. , Agilent SureSelectTM, Illumina TruSeq CaptureTM, and Nimblegen SeqCap EZ ChoiceTM).
  • kits designed for this purpose e.g. , Agilent SureSelectTM, Illumina TruSeq CaptureTM, and Nimblegen SeqCap EZ ChoiceTM.
  • genomic DNA containing the loci to be analyzed can be hybridized to biotinylated capture RNA fragments to form biotinylated RNA/genomic DNA complexes.
  • DNA capture probes may be utilized resulting in the formation of biotinylated DNA/genomic DNA hybrids.
  • Streptavidin coated magnetic beads and a magnetic force can be used to separate the biotinylated RNA/genomic DNA complexes from those genomic DNA fragments not present within a biotinylated RNA/genomic DNA complex.
  • the obtained biotinylated RNA/genomic DNA complexes can be treated to remove the captured RNA from the magnetic beads, thereby leaving intact genomic DNA fragments containing a locus to be analyzed.
  • These intact genomic DNA fragments containing the loci to be analyzed can be amplified using, for example, PCR techniques.
  • the amplified genomic DNA fragments can be sequenced using a high-throughput sequencing technology or a next-generation sequencing technology such as Illumina HiSeqTM, Illumina MiSeqTM, Life Technologies SoLIDTM or Ion TorrentTM, or Roche 454TM.
  • a high-throughput sequencing technology or a next-generation sequencing technology such as Illumina HiSeqTM, Illumina MiSeqTM, Life Technologies SoLIDTM or Ion TorrentTM, or Roche 454TM.
  • the sequencing results from the genomic DNA fragments can be used to identify loci as exhibiting or not exhibiting a CA, analogous to the microarray analysis described herein.
  • an analysis of the genotype of loci over a length of a chromosome can be performed to determine the length of CA Regions. For example, a stretch of SNP locations that are spaced apart ( e.g., spaced about 25 kb to about 100 kb apart) along a chromosome can be evaluated by sequencing, and the sequencing results used to determine not only the presence of a CA Region but also the length of that CA Region. Obtained sequencing results can be used to generate a graph that plots allele dosages along a chromosome.
  • a selection process can be used to select loci (e.g., SNP loci) to be evaluated using an assay configured to genotype loci (e.g., SNP array-based assays and sequencing-based assays). For example, any human SNP location can be selected for inclusion in a SNP array-based assay or a sequencing-based assay configured to genotype loci.
  • 0.5, 1.0, 1.5, 2.0, 2.5 million or more SNP locations present within the human genome can be evaluated to identify those SNPs that (a) are not present on the Y chromosome, (b) are not mitochondrial SNPs, (c) have a minor allele frequency of at least about five percent in Caucasians, (d) have a minor allele frequency of at least about one percent in three races other than Caucasians ( e.g. , Chinese, Japanese, and Yoruba), and/or (e) do not have a significant deviation from Hardy Weinberg equilibrium in any of the four races. In some cases, more than 100,000, 150,000, or 200,000 human SNPs can be selected that meet criteria (a) through (e).
  • a group of SNPs e.g., top 110,000 SNPs
  • the SNPs have a high degree of allele frequency in Caucasians, cover the human genome in a somewhat evenly spaced manner (e.g. , at least one SNP every about 25 kb to about 500 kb), and are not in linkage disequilibrium with another selected SNP for in any of the four races.
  • about 40, 50, 60, 70, 80, 90, 100, 110, 120, 130 thousand or more SNPs can be selected as meeting each of these criteria and included in an assay configured to identify CA Regions across a human genome.
  • SNPs can be selected for analysis with a SNP array-based assay, and between about 45,000 and about 55,000 ( e.g., about 54,000) SNPs can be selected for analysis with a sequencing-based assay.
  • any appropriate type of sample can be assessed.
  • a sample containing cancer cells can be assessed to determine if the genome of the cancer cells contains an HRD signature, lacks an HRD signature, has an increased number of Indicator CA Regions or has an increased CA Region Score.
  • samples containing cancer cells that can be assessed as described herein include, without limitation, tumor biopsy samples (e.g. , breast tumor biopsy samples), formalin-fixed, paraffin-embedded tissue samples containing cancer cells, core needle biopsies, fine needle aspirates, and samples containing cancer cells shed from a tumor ( e.g., blood, urine or other bodily fluids).
  • the sample can be prepared by DNA extraction using a genomic DNA extraction kit optimized for FFPE tissue, including but not limited to those described above (e.g. , QuickExtractTM FFPE DNA Extraction Kit (EpicentreTM), and QIAampTM DNA FFPE Tissue Kit (QiagenTM)).
  • a genomic DNA extraction kit optimized for FFPE tissue including but not limited to those described above (e.g. , QuickExtractTM FFPE DNA Extraction Kit (EpicentreTM), and QIAampTM DNA FFPE Tissue Kit (QiagenTM)).
  • laser dissection techniques can be performed on a tissue sample to minimize the number of non-cancer cells within a cancer cell sample to be assessed.
  • antibody based purification methods can be used to enrich for cancer cells and/or deplete non-cancer cells.
  • antibodies that could be used for cancer cell enrichment include, without limitation, anti-EpCAM, anti-TROP-2, anti-c-Met, anti-Folate binding protein, anti-N-Cadherin, anti-CD318, anti-antimesencymal stem cell antigen, anti-Her2, anti-MUCl, anti-EGFR, anti-cytokeratins ( e.g. , cytokeratin 7, cytokeratin 20, etc.), anti-Caveolin-1, anti-PSA, anti-CA125, and anti-surfactant protein antibodies.
  • cancer cell can be assessed using the methods and materials described herein.
  • breast cancer cells, ovarian cancer cells, liver cancer cells, esophageal cancer cells, lung cancer cells, head and neck cancer cells, prostate cancer cells, colon, rectal, or colorectal cancer cells, and pancreatic cancer cells can be assessed to determine if the genome of the cancer cells contains an HRD signature, lacks an HRD signature, has an increased number of Indicator CA Regions or has an increased CA Region Score.
  • the cancer cells are primary or metastatic cancer cells of ovarian cancer, breast cancer, lung cancer or esophageal cancer.
  • one or more e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, or 23
  • pairs of chromosomes can be assessed.
  • the genome of cancer cells is assessed for the presence or absence of an HRD signature using one or more ( e.g., one, two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23) pairs of chromosomes.
  • a pair to be assessed can include the pair of X sex chromosomes; whereas, in the case of males, a pair of any autosomal chromosomes (i.e., any pair other than the pair of X and Y sex chromosomes) can be assessed.
  • the chromosome number 17 pair may be excluded from the analysis. It has been determined that certain chromosomes carry unusually high levels of CA in certain cancers and, thus, it can be helpful to exclude such chromosomes when analyzing samples as described herein from patients having these cancers. In some cases, the sample is from a patient having ovarian cancer, and the chromosome to be excluded is chromosome 17.
  • a predefined number of chromosomes may be analyzed to determine the number of Indicator CA Regions (or the CA Region Score or Combined CA Region Score), preferably the number of CA Regions of a length of greater than 9 megabases, 10 megabases, 12 megabases, 14 megabases, more preferably greater than 15 megabases.
  • the sizes of all identified Indicator CA Regions may be summed up to obtain a total length of Indicator CA Regions.
  • patients having cancer cells (or samples derived therefrom) identified as having an HRD signature status can be classified, based at least in part on such HRD signature, as being likely to respond to a particular cancer treatment regimen.
  • patients having cancer cells with an HRD signature can be classified, based at least in part on such HRD signature, as being likely to respond to a cancer treatment regimen that includes the use of a DNA damaging agent, a synthetic lethality agent (e.g., a PARP inhibitor), radiation, or a combination thereof.
  • the patients are treatment naive patients.
  • DNA damaging agents include, without limitation, platinum-based chemotherapy drugs (e.g.
  • Synthetic lethality therapeutic approaches typically involve administering an agent that inhibits at least one critical component of a biological pathway that is especially important to a particular tumor cell's survival. For example, when a tumor cell has a deficient homologous repair pathway (e.g.
  • inhibitors of poly ADP ribose polymerase can be especially potent against such tumors because two pathways critical to survival become obstructed (one biologically, e.g. , by BRCA1 mutation, and the other synthetically, e.g. , by administration of a pathway drug).
  • Synthetic lethality approaches to cancer therapy are described in, e.g., O'Brien et al., Converting cancer mutations into therapeutic opportunities, EMBO MOL. MED. (2009) 1:297-299 .
  • Examples of synthetic lethality agents include, without limitation, PARP inhibitors or double strand break repair inhibitors in homologous repair-deficient tumor cells, PARP inhibitors in PTEN-deficient tumor cells, methotrexate in MSH2-deficient tumor cells, etc.
  • Examples of PARP inhibitors include, without limitation, olaparib, iniparib, and veliparib.
  • Examples of double strand break repair inhibitors include, without limitation, KU55933 (ATM inhibitor) and NU7441 (DNA-PKcs inhibitor).
  • Examples of information that can be used in addition to the presence of an HRD signature to base a classification of being likely to respond to a particular cancer treatment regimen include, without limitation, previous treatment results, germline or somatic DNA mutations, gene or protein expression profiling (e.g., ER/PR/HER2 status, PSA levels), tumor histology (e.g., adenocarcinoma, squamous cell carcinoma, papillary serous carcinoma, mucinous carcinoma, invasive ductal carcinoma, ductal carcinoma in situ (non-invasive), etc.), disease stage, tumor or cancer grade (e.g., well, moderately, or poorly differentiated ( e.g. , Gleason, modified Bloom Richardson), etc.), number of previous courses of treatment, etc.
  • gene or protein expression profiling e.g., ER/PR/HER2 status, PSA levels
  • tumor histology e.g., adenocarcinoma, squamous cell carcinoma, papillary serous carcinoma,
  • the cancer patient can be treated with such a cancer treatment regimen.
  • the patients are treatment naive patients.
  • the invention thus provides a method of treating a patient comprising detecting an HRD signature as described herein and administering (or recommending or prescribing) a treatment regimen comprising the use of a DNA damaging agent, a PARP inhibitor, radiation, or a combination thereof. Any appropriate method for treating the cancer at issue can be used to treat a cancer patient identified as having cancer cells having an HRD signature.
  • platinum-based chemotherapy drugs or a combination of platinum-based chemotherapy drugs can be used to treat cancer as described elsewhere (see, e.g., U.S. Patent Nos. 3,892,790 , 3,904,663 , 7,759,510 , 7,759,488 and 7,754,684 .
  • anthracyclines or a combination of anthracyclines can be used to treat cancer as described elsewhere ( see, e.g., U.S. Patent Nos. 3,590,028 , 4,138,480 , 4,950,738 , 6,087,340 , 7,868,040 , and 7,485,707 ).
  • topoisomerase I inhibitors or a combination of topoisomerase I inhibitors can be used to treat cancer as described elsewhere ( see, e.g., U.S. Patent Nos. 5,633,016 and 6,403,563 .
  • PARP inhibitors or a combination of PARP inhibitors can be used to treat cancer as described elsewhere ( see, e.g., U.S. Patent Nos. 5,177,075 , 7,915,280 , and 7,351,701 .
  • radiation can be used to treat cancer as described elsewhere ( see, e.g., U.S. Patent No. 5,295,944 ).
  • a combination comprising different agents (e.g., a combination comprising any of platinum-based chemotherapy drugs, anthracyclines, topoisomerase I inhibitors, and/or PARP inhibitors) with or without radiation treatments can be used to treat cancer.
  • a combination treatment may comprise any of the above agents or treatments (e.g. , a DNA damaging agent, a PARP inhibitor, radiation, or a combination thereof) together with another agent or treatment- e.g ., a taxane agent (e.g. , doxetaxel, paclitaxel, abraxane), a growth factor or growth factor receptor inhibitor (e.g.
  • erlotinib e.g. , 5-flourouracil, methotrexate.
  • an antimetabolite e.g. , 5-flourouracil, methotrexate
  • patients identified as having cancer cells lacking an HRD signature can be classified, based at least in part on a sample lacking an HRD signature, as being less likely to respond to a treatment regimen that includes a DNA damaging agent, a PARP inhibitor, radiation, or a combination thereof.
  • a patient can be classified as likely to respond to a cancer treatment regimen that includes the use of one or more cancer treatment agents not associated with HDR, such as a taxane agent (e.g. , doxetaxel, paclitaxel, abraxane), a growth factor or growth factor receptor inhibitor (e.g.
  • the patients are treatment naive patients. Once classified as being likely to respond to a particular cancer treatment regimen (e.g., a cancer treatment regimen that includes the use of a cancer treatment agent not associated with HDR), the cancer patient can be treated with such a cancer treatment regimen.
  • a cancer treatment regimen e.g., a cancer treatment regimen that includes the use of a cancer treatment agent not associated with HDR
  • the invention thus provides a method of treating a patient comprising detecting the absence of an HRD signature as described herein and administering (or recommending or prescribing) a treatment regimen not comprising the use of a DNA damaging agent, a PARP inhibitor, radiation, or a combination thereof.
  • the treatment regimen comprises one or more of a taxane agent (e.g. , doxetaxel, paclitaxel, abraxane), a growth factor or growth factor receptor inhibitor (e.g.
  • any appropriate method for the cancer being treated can be used to treat a cancer patient identified as having cancer cells lacking an HRD signature.
  • information that can be used in addition to the absence of an HRD signature to base a classification of being likely to respond to a particular cancer treatment regimen include, without limitation, previous treatment results, germline or somatic DNA mutations, gene or protein expression profiling (e.g.
  • tumor histology e.g. , adenocarcinoma, squamous cell carcinoma, papillary serous carcinoma, mucinous carcinoma, invasive ductal carcinoma, ductal carcinoma in situ (non-invasive), etc.
  • disease stage e.g., well, moderately, or poorly differentiated ( e.g. , Gleason, modified Bloom Richardson), etc.), number of previous courses of treatment, etc.
  • the patient can be assessed to determine whether or not the treatment regimen has an effect. If a beneficial effect is detected, the patient can continue with the same or a similar cancer treatment regimen. If a minimal or no beneficial effect is detected, then adjustments to the cancer treatment regimen can be made. For example, the dose, frequency of administration, or duration of treatment can be increased. In some cases, additional anti-cancer agents can be added to the treatment regimen or a particular anti-cancer agent can be replaced with one or more different anti-cancer agents. The patient being treated can continue to be monitored as appropriate, and changes can be made to the cancer treatment regimen as appropriate.
  • an HRD signature can be used to determine a patient's prognosis.
  • this document features a method for determining a patient's prognosis based at least in part of detecting the presence or absence of an HRD signature in a sample from the patient.
  • the method comprises, or consists essentially of, (a) determining whether a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature as described herein ( e.g.
  • Prognosis may include the patient's likelihood of survival (e.g. , progression-free survival, overall survival), wherein a relatively good prognosis would include an increased likelihood of survival as compared to some reference population ( e.g. , average patient with this patient's cancer type/subtype, average patient not having an HRD signature, etc.). Conversely, a relatively poor prognosis in terms of survival would include a decreased likelihood of survival as compared to some reference population ( e.g. , average patient with this patient's cancer type/subtype, average patient having an HRD signature, etc.).
  • this document provides methods for assessing patients for cells (e.g., cancer cells) having an HRD signature.
  • one or more clinicians or medical professionals can determine whether a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature.
  • one or more clinicians or medical professionals can determine if a patient contains cancer cells having an HRD signature by obtaining a cancer cell sample from the patient and assessing the DNA of cancer cells of the cancer cell sample to determine the presence or absence of an HRD signature as described herein.
  • one or more clinicians or medical professionals can obtain a cancer cell sample from a patient and provide that sample to a testing laboratory having the ability to assess DNA of cancer cells of the cancer cell sample to provide an indication about the presence or absence of an HRD signature as described herein.
  • the patients are treatment naive patients.
  • the one or more clinicians or medical professionals can determine if a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature by receiving information about the presence or absence of an HRD signature as described herein directly or indirectly from the testing laboratory.
  • a testing laboratory after assessing DNA of cancer cells for presence or absence of an HRD signature as described herein, can provide a clinician or medical professional with, or access to, a written, electronic, or oral report or medical record that provides an indication about the presence or absence of an HRD signature for a particular patient (or patient sample) being assessed.
  • a written, electronic, or oral report or medical record can allow the one or more clinicians or medical professionals to determine if a particular patient being assessed contains cancer cells having an HRD signature.
  • a clinician or medical professional or group of clinicians or medical professionals determines that a particular patient being assessed contains cancer cells having an HRD signature
  • the clinician or medical professional can classify that patient as having cancer cells whose genome contains the presence of an HRD signature.
  • the patients are treatment naive patients.
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells whose genome contains the presence of an HRD signature as having cancer cells deficient in (or likely to be deficient in) HDR.
  • Such a diagnosis can be based solely on a determination that a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature or can be based at least in part on a determination that a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature.
  • a patient determined to have cancer cells having an HRD signature can be diagnosed as likely to be deficient in HDR based on the combination of the presence of an HRD signature and deficient status in one or more tumor suppressor genes (e.g. , BRCA1/2, RAD51C), a family history of cancer, or the presence of behavioral risk factors ( e.g. , smoking).
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells whose genome contains the presence of an HRD signature as having cancer cells likely to contain genetic mutations in one or more genes in the HDR pathway.
  • the patients are treatment naive patients.
  • Such a diagnosis can be based solely on a determination that a particular patient being assessed contains cancer cells having a genome containing an HRD signature or can be based at least in part on a determination that a particular patient being assessed contains cancer cells having a genome containing an HRD signature.
  • a patient determined to have cancer cells whose genome contains the presence of an HRD signature can be diagnosed as having cancer cells likely to contain genetic mutations in one or more genes in the HDR pathway based on the combination of the presence of an HRD signature and a family history of cancer, or the presence of behavioral risk factors (e.g. , smoking).
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells having an HRD signature as having cancer cells likely to respond to a particular cancer treatment regimen.
  • the patients are treatment naive patients.
  • Such a diagnosis can be based solely on a determination that a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature or can be based at least in part on a determination that a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having an HRD signature.
  • a patient determined to have cancer cells having an HRD signature can be diagnosed as being likely to respond to a particular cancer treatment regimen based on the combination of the presence of an HRD signature and deficient status in one or more tumor suppressor genes (e.g., BRCA1/2, RAD51), a family history of cancer, or the presence of behavioral risk factors (e.g. , smoking).
  • tumor suppressor genes e.g., BRCA1/2, RAD51
  • a family history of cancer e.g., a family history of cancer
  • behavioral risk factors e.g. , smoking
  • a patient determined to have cancer cells having an HRD signature can be diagnosed as likely to respond to a cancer treatment regimen that includes the use of a platinum-based chemotherapy drug such as cisplatin, carboplatin, oxaliplatin, or picoplatin, an anthracycline such as epirubicin or doxorubicin, a topoisomerase I inhibitor such as campothecin, topotecan, or irinotecan, a PARP inhibitor, radiation, a combination thereof, or a combination of any of the preceding with another anti-cancer agent.
  • a platinum-based chemotherapy drug such as cisplatin, carboplatin, oxaliplatin, or picoplatin
  • an anthracycline such as epirubicin or doxorubicin
  • a topoisomerase I inhibitor such as campothecin, topotecan, or irinotecan
  • PARP inhibitor radiation, a combination thereof, or a combination of any of the preceding with
  • a clinician or medical professional or group of clinicians or medical professionals determines that a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) having a genome lacking an HRD signature
  • the clinician or medical professional can classify that patient as having cancer cells whose genome lacks an HRD signature.
  • the patients are treatment naive patients.
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells containing a genome lacking an HRD signature as having cancer cells likely to have functional HDR.
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells containing a genome lacking an HRD signature as having cancer cells that do not likely contain genetic mutations in one or more genes in the HDR pathway.
  • a clinician or medical professional or group of clinicians or medical professionals can diagnose a patient determined to have cancer cells containing a genome lacking an HRD signature or containing an increased number of CA Regions that cover the whole chromosome as having cancer cells that are less likely to respond to a platinum-based chemotherapy drug such as cisplatin, carboplatin, oxalaplatin, or picoplatin, an anthracycline such as epirubincin or doxorubicin, a topoisomerase I inhibitor such as campothecin, topotecan, or irinotecan, a PARP inhibitor, or radiation and/or more likely to respond to a cancer treatment regimen that includes the use of a cancer treatment agent not associated with HDR such as one or more taxane
  • this document also provides methods for performing a diagnostic analysis of a nucleic acid sample (e.g. , a genomic nucleic acid sample or nucleic acids amplified therefrom) of a cancer patient to determine if a sample from the patient comprises cancer cells (or whether a sample comprises DNA derived from such cells) containing an HRD signature and/or an increased number of CA Regions that cover the whole chromosome.
  • a nucleic acid sample e.g. , a genomic nucleic acid sample or nucleic acids amplified therefrom
  • the patients are treatment naive patients.
  • one or more laboratory technicians or laboratory professionals can detect the presence or absence of an HRD signature in the genome of cancer cells (or DNA derived therefrom) of the patient or the presence or absence of an increased number of CA Regions that cover the whole chromosome in the genome of cancer cells of the patient.
  • one or more laboratory technicians or laboratory professionals can detect the presence or absence of an HRD signature or the presence or absence of an increased number of CA Regions that cover the whole chromosome in the genome of cancer cells of the patient by (a) receiving a cancer cell sample obtained from the patient, receiving a genomic nucleic acid sample obtained from cancer cells obtained from the patient, or receiving a sample containing nucleic acids enriched and/or amplified from such a genomic nucleic acid sample obtained from cancer cells obtained from the patient and (b) performing an analysis (e.g.
  • a SNP array-based assay or a sequencing-based assay using the received material to detect the presence or absence of an HRD signature or the presence or absence of an increased number of CA Regions that cover the whole chromosome as described herein.
  • one or more laboratory technicians or laboratory professionals can receive a sample to be analyzed (e.g., a cancer cell sample obtained from the patient, a genomic nucleic acid sample obtained from cancer cells obtained from the patient, or a sample containing nucleic acids enriched and/or amplified from such a genomic nucleic acid sample obtained from cancer cells obtained from the patient) directly or indirectly from a clinician or medical professional.
  • the patients are treatment naive patients.
  • the laboratory technician or laboratory professional can associate that HRD signature or the result (or results or a summary of results) of the performed diagnostic analysis with the corresponding patient's name, medical record, symbolic/numerical identifier, or a combination thereof.
  • identification can be based solely on detecting the presence of an HRD signature or can be based at least in part on detecting the presence of an HRD signature.
  • a laboratory technician or laboratory professional can identify a patient having cancer cells that were detected to have an HRD signature as having cancer cells potentially deficient in HDR (or as having an increased likelihood of responding to a particular treatment as described at length herein) based on a combination of the presence of an HRD signature and the results of other genetic and biochemical tests performed at the testing laboratory.
  • the patients are treatment naive patients.
  • the laboratory technician or laboratory professional can associate the absence of an HRD signature or the result (or results or a summary of results) of the performed diagnostic analysis with the corresponding patient's name, medical record, symbolic/numerical identifier, or a combination thereof.
  • a laboratory technician or laboratory professional or group of laboratory technicians or laboratory professionals can identify a patient having cancer cells that were detected to lack an HRD signature as having cancer cells with potentially intact HDR (or having a decreased likelihood of responding to a particular treatment as described at length herein) either based solely on the absence of an HRD signature or based on a combination of the presence of an HRD signature and the results of other genetic and biochemical tests performed at the testing laboratory.
  • the patients are treatment naive patients.
  • results of any analyses according to the invention will often be communicated to physicians, genetic counselors and/or patients (or other interested parties such as researchers) in a transmittable form that can be communicated or transmitted to any of the above parties.
  • a form can vary and can be tangible or intangible.
  • the results can be embodied in descriptive statements, diagrams, photographs, charts, images or any other visual forms. For example, graphs or diagrams showing genotype or LOH (or HRD status) information can be used in explaining the results.
  • the statements and visual forms can be recorded on a tangible medium such as papers, computer readable media such as floppy disks, compact disks, flash memory, etc., or in an intangible medium, e.g. , an electronic medium in the form of email or website on internet or intranet.
  • results can also be recorded in a sound form and transmitted through any suitable medium, e.g. , analog or digital cable lines, fiber optic cables, etc., via telephone, facsimile, wireless mobile phone, internet phone and the like.
  • the information and data on a test result can be produced anywhere in the world and transmitted to a different location.
  • the information and data on a test result may be generated, cast in a transmittable form as described above, and then imported into the United States.
  • the present invention also encompasses a method for producing a transmittable form of information on an HRD signature for at least one patient sample.
  • the method comprises the steps of (1) determining an HRD signature according to methods of the present invention; and (2) embodying the result of the determining step in a transmittable form.
  • the transmittable form is a product of such a method.
  • an HRD signature e.g. , the total number of Indicator CA Regions or a CA Region Score or Combined CA Region Score greater than a reference
  • a particular clinical feature e.g. , an increased likelihood of a deficiency in the BRCA1 or BRCA2 gene; an increased likelihood of HDR deficiency; an increased likelihood of response to a treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor; etc.
  • optionally correlating the absence of a HRD signature to one or more other clinical features.
  • another embodiment of the invention may involve, in addition to or instead of a correlating step, one or both of the following steps: (a) concluding that the patient has the clinical feature based at least in part on the presence or absence of the HRD signature; or (b) communicating that the patient has the clinical feature based at least in part on the presence or absence of the HRD signature.
  • one embodiment described in this document is a method of predicting a cancer patient's response to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor, said method comprising: (1) determining in a sample two or more of (a) an LOH Region Score for the sample; (b) a TAI Region Score for the sample; or (c) an LST Region Score for the sample; and (2)(a) correlating a combination of two or more of the LOH Region Score, the TAI Region Score and the LST Region Score ( e.g., a Combined CA Region Score) exceeding a reference to an increased likelihood of responding to the treatment regimen; or optionally (2)(b) correlating a combination of two or more of the LOH Region Score, the TAI Region Score and the LST Region Score ( e.g., a Combined CA Region Score) not exceeding a reference to a not increased likelihood of responding to the treatment regimen;
  • One such embodiment provides a method of predicting a cancer patient's response to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor, said method comprising: (1) determining in a sample two or more of (a) an LOH Region Score for the sample; (b) a TAI Region Score for the sample; or (c) an LST Region Score for the sample; or (d) an average ( e.g.
  • Another such embodiment provides a method of predicting a cancer patient's response to a cancer treatment regimen comprising a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor, said method comprising: (1) determining in a sample two or more of (a) an LOH Region Score for the sample; (b) a TAI Region Score for the sample; or (c) an LST Region Score for the sample; or (d) an average ( e.g.
  • arithmetic mean of the LOH Region Score, the TAI Region Score, and the LST Region Score, exceeding a reference; or optionally (2)(b) communicating that said patient has a not increased likelihood of responding to said cancer treatment regimen based at least in part on a combination of two or more of the LOH Region Score, the TAI Region Score and the LST Region Score (e.g., a Combined CA Region Score); or an average (e.g., arithmetic mean) of the LOH Region Score, the TAI Region Score, and the LST Region Score, not exceeding a reference.
  • a combination of two or more of the LOH Region Score, the TAI Region Score and the LST Region Score e.g., a Combined CA Region Score
  • an average e.g., arithmetic mean
  • such correlating, concluding or communicating may comprise assigning a risk or likelihood of the clinical feature occurring based at least in part on the particular assay or analysis output.
  • risk is a percentage probability of the event or outcome occurring.
  • the patient is assigned to a risk group (e.g. , low risk, intermediate risk, high risk, etc.).
  • low risk is any percentage probability below 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50%.
  • intermediate risk is any percentage probability above 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% and below 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, or 75%.
  • high risk is any percentage probability above 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 99%.
  • communicating means to make such information known to another person or transfer such information to a thing (e.g., a computer).
  • a patient's prognosis or likelihood of response to a particular treatment is communicated.
  • the information used to arrive at such a prognosis or response prediction e.g. , HRD signature according to the present invention, etc.
  • This communication may be auditory ( e.g., verbal), visual ( e.g., written), electronic ( e.g., data transferred from one computer system to another), etc.
  • communicating a cancer classification e.g.
  • the cancer classification comprises generating a report that communicates the cancer classification.
  • the report is a paper report, an auditory report, or an electronic record.
  • the report is displayed and/or stored on a computing device ( e.g. , handheld device, desktop computer, smart device, website, etc.).
  • the cancer classification is communicated to a physician ( e.g. , a report communicating the classification is provided to the physician).
  • the cancer classification is communicated to a patient (e.g., a report communicating the classification is provided to the patient). Communicating a cancer classification can also be accomplished by transferring information (e.g.
  • data embodying the classification to a server computer and allowing an intermediary or end-user to access such information (e.g. , by viewing the information as displayed from the server, by downloading the information in the form of one or more files transferred from the server to the intermediary or end-user's device, etc.).
  • an embodiment of the invention comprises concluding some fact (e.g., a patient's prognosis or a patient's likelihood of response to a particular treatment regimen)
  • this may include in some embodiments a computer program concluding such fact, typically after performing an algorithm that applies information on CA Regions according to the present invention.
  • the present invention encompasses a related embodiment involving a test value or score (e.g., CA Region Score, LOH Region Score, etc.) derived from, incorporating, and/or, at least to some degree, reflecting such number or length.
  • a test value or score e.g., CA Region Score, LOH Region Score, etc.
  • the bare CA Region numbers or lengths need not be used in the various methods, systems, etc. of the invention; a test value or score derived from such numbers or lengths may be used.
  • one embodiment of the invention provides a method of treating cancer in a patient, comprising: (1) determining in a sample from said patient two or more of, or an average (e.g. , arithmetic mean) of, (a) the number of Indicator LOH Regions, (b) the number of Indicator TAI Regions, or (c) the number of Indicator LST Regions; (2) providing one or more test values derived from said number of Indicator LOH Regions, Indicator TAI Regions, and/or Indicator LST Regions; (3) comparing said test value(s) to one or more reference values (e.g.
  • test values derived from the number of Indicator LOH regions, Indicator TAI Regions, and/or Indicator LST Regions in a reference population e.g. , mean, median, terciles, quartiles, quintiles, etc.
  • administering or recommending or prescribing or initiating a treatment regimen comprising chemotherapy and/or a synthetic lethality agent based at least in part on said comparing step revealing that one or more of the test values is greater ( e.g., at least 2-, 3-, 4-, 5-, 6-, 7-, 8-, 9-, or 10-fold greater; at least 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 standard deviations greater) than at least one said reference value; or optionally (4)(b) recommending or prescribing or initiating a treatment regimen not comprising chemotherapy and/or a synthetic lethality agent based at least in part on said comparing step revealing that one or more of the test values
  • the invention encompasses, mutatis mutandis, corresponding embodiments where the test value or score is used to determine the patient's prognosis, the patient's likelihood of response to a particular treatment regimen, the patient's or patient's sample's likelihood of having a BRCA1, BRCA2, RAD51C or HDR deficiency, etc.
  • Figure 8 shows an exemplary process by which a computing system (or a computer program ( e.g. , software) containing computer-executable instructions) can identify LOH loci or regions from genotype data as described herein.
  • This process may be adapted to use in determining TAI and LST as will be apparent to those skilled in the art. If the observed ratio of the signals of two alleles, A and B, is two to one, there are two possibilities. The first possibility is that cancer cells have LOH with deletion of allele B in a sample with 50% contamination with normal cells. The second possibility is that there is no LOH but allele A is duplicated in a sample with no contamination with normal cells.
  • the process begins at box 1500, where the following data are collected by the computing system; (1) sample-specific normalized signal intensities for both alleles of each locus and (2) assay-specific (specific for different SNP arrays and for sequence based approach) set of parameters defined based on analysis of large number of samples with known ASCN profiles.
  • any appropriate assay such as a SNP array-based assay or sequencing-based assay can be used to assess loci along a chromosome for homozygosity or heterozygosity.
  • a system including a signal detector and a computer can be used to collect data (e.g. , fluorescent signals or sequencing results) regarding the homozygous or heterozygous nature of the plurality of loci (e.g.
  • ASCN allele specific copy numbers
  • SNP allele specific copy numbers
  • a likelihood function is used to determine whether a homozygous locus or region of homozygous loci is due to LOH. This can be conceptually analogous to a previously described algorithm designed to reconstruct total copy number (rather than ASCN) at each locus (e.g., SNP). See International Application No. PCT/US2011/026098 to Abkevich et al.
  • the likelihood function can be maximized over ASCN of all loci, level of contamination with benign tissue, total copy number averaged over the whole genome, and sample specific noise level.
  • an LOH region is determined as a stretch of SNPs with one of the ASCNs (paternal or maternal) being zero.
  • the computer process further comprises a step of inquiring or determining whether a patient is treatment naive.
  • Figure 3 shows an exemplary process by which a computing system can determine the presence or absence of an LOH signature and is included to illustrate how this process can, as will be apparent to those skilled in the art, be applied to TAI and LST.
  • the process begins at box 300, where data regarding the homozygous or heterozygous nature of a plurality of loci along a chromosome is collected by the computing system.
  • any appropriate assay such as a SNP array-based assay or sequencing-based assay can be used to assess loci along a chromosome for homozygosity or heterozygosity.
  • a system including a signal detector and a computer can be used to collect data (e.g.
  • data regarding the homozygous or heterozygous nature of a plurality of loci as well as the location or spatial relationship of each locus is assessed by the computing system to determine the length of any LOH regions present along a chromosome.
  • data regarding the number of LOH regions detected and the length of each detected LOH region is assessed by the computing system to determine the number of LOH regions that have a length (a) greater than or equal to a preset number of Mb ( e.g., 15 Mb) and (b) less than the entire length of the chromosome containing that LOH region.
  • the computing system can determine the total or combined LOH length as described above.
  • the computing system formats an output providing an indication of the presence or absence of an HRD signature.
  • the computing system can present the output to a user (e.g., a laboratory technician, clinician, or medical professional).
  • a user e.g., a laboratory technician, clinician, or medical professional.
  • the presence or absence of an HRD signature can be used to provide an indication about a patient's likely HDR status, an indication about the likely presence or absence of genetic mutations in genes of the HDR pathway, and/or an indication about possible cancer treatment regimens.
  • FIG. 4 is a diagram of an example of a computer device 1400 and a mobile computer device 1450, which may be used with the techniques described herein.
  • Computing device 1400 is intended to represent various forms of digital computers, such as laptops, desktops, workstations, personal digital assistants, servers, blade servers, mainframes, and other appropriate computers.
  • Computing device 1450 is intended to represent various forms of mobile devices, such as personal digital assistants, cellular telephones, smart phones, and other similar computing devices.
  • the components shown here, their connections and relationships, and their functions, are meant to be exemplary only, and are not meant to limit implementations of the inventions described and/or claimed in this document.
  • Computing device 1400 includes a processor 1402, memory 1404, a storage device 1406, a high-speed interface 1408 connecting to memory 1404 and high-speed expansion ports 1410, and a low speed interface 1415 connecting to low speed bus 1414 and storage device 1406.
  • Each of the components 1402, 1404, 1406, 1408, 1410, and 1415 are interconnected using various busses, and may be mounted on a common motherboard or in other manners as appropriate.
  • the processor 1402 can process instructions for execution within the computing device 1400, including instructions stored in the memory 1404 or on the storage device 1406 to display graphical information for a GUI on an external input/output device, such as display 1416 coupled to high speed interface 1408.
  • multiple processors and/or multiple buses may be used, as appropriate, along with multiple memories and types of memory.
  • multiple computing devices 1400 may be connected, with each device providing portions of the necessary operations (e.g., as a server bank, a group of blade servers, or a multi-processor system).
  • the memory 1404 stores information within the computing device 1400.
  • the memory 1404 is a volatile memory unit or units.
  • the memory 1404 is a non-volatile memory unit or units.
  • the memory 1404 may also be another form of computer-readable medium, such as a magnetic or optical disk.
  • the storage device 1406 is capable of providing mass storage for the computing device 1400.
  • the storage device 1406 may be or contain a computer-readable medium, such as a floppy disk device, a hard disk device, an optical disk device, or a tape device, a flash memory or other similar solid state memory device, or an array of devices, including devices in a storage area network or other configurations.
  • a computer program product can be tangibly embodied in an information carrier.
  • the computer program product may also contain instructions that, when executed, perform one or more methods, such as those described herein.
  • the information carrier is a computer- or machine-readable medium, such as the memory 1404, the storage device 1406, memory on processor 1402, or a propagated signal.
  • the high speed controller 1408 manages bandwidth-intensive operations for the computing device 1400, while the low speed controller 1415 manages lower bandwidth-intensive operations.
  • the high-speed controller 1408 is coupled to memory 1404, display 1416 ( e.g., through a graphics processor or accelerator), and to high-speed expansion ports 1410, which may accept various expansion cards (not shown).
  • the low-speed controller 1415 is coupled to storage device 1406 and low-speed expansion port 1414.
  • the low-speed expansion port which may include various communication ports (e.g.
  • USB Universal Serial Bus
  • Ethernet Ethernet
  • wireless Ethernet may be coupled to one or more input/output devices, such as a keyboard, a pointing device, a scanner, an optical reader, a fluorescent signal detector, or a networking device such as a switch or router, e.g. , through a network adapter.
  • input/output devices such as a keyboard, a pointing device, a scanner, an optical reader, a fluorescent signal detector, or a networking device such as a switch or router, e.g. , through a network adapter.
  • the computing device 1400 may be implemented in a number of different forms, as shown in the figure. For example, it may be implemented as a standard server 1420, or multiple times in a group of such servers. It may also be implemented as part of a rack server system 1424. In addition, it may be implemented in a personal computer such as a laptop computer 1422. Alternatively, components from computing device 1400 may be combined with other components in a mobile device (not shown), such as device 1450. Each of such devices may contain one or more of computing device 1400, 1450, and an entire system may be made up of multiple computing devices 1400, 1450 communicating with each other.
  • Computing device 1450 includes a processor 1452, memory 1464, an input/output device such as a display 1454, a communication interface 1466, and a transceiver 1468, among other components (e.g ., a scanner, an optical reader, a fluorescent signal detector).
  • the device 1450 may also be provided with a storage device, such as a microdrive or other device, to provide additional storage.
  • a storage device such as a microdrive or other device, to provide additional storage.
  • Each of the components 1450, 1452, 1464, 1454, 1466, and 1468 are interconnected using various buses, and several of the components may be mounted on a common motherboard or in other manners as appropriate.
  • the processor 1452 can execute instructions within the computing device 1450, including instructions stored in the memory 1464.
  • the processor may be implemented as a chipset of chips that include separate and multiple analog and digital processors.
  • the processor may provide, for example, for coordination of the other components of the device 1450, such as control of user interfaces, applications run by device 1450, and wireless communication by device 1450.
  • Processor 1452 may communicate with a user through control interface 1458 and display interface 1456 coupled to a display 1454.
  • the display 1454 may be, for example, a TFT LCD (Thin-Film-Transistor Liquid Crystal Display) or an OLED (Organic Light Emitting Diode) display, or other appropriate display technology.
  • the display interface 1456 may comprise appropriate circuitry for driving the display 1454 to present graphical and other information to a user.
  • the control interface 1458 may receive commands from a user and convert them for submission to the processor 1452.
  • an external interface 1462 may be provide in communication with processor 1452, so as to enable near area communication of device 1450 with other devices. External interface 1462 may provide, for example, for wired communication in some implementations, or for wireless communication in other implementations, and multiple interfaces may also be used.
  • the memory 1464 stores information within the computing device 1450.
  • the memory 1464 can be implemented as one or more of a computer-readable medium or media, a volatile memory unit or units, or a non-volatile memory unit or units.
  • Expansion memory 1474 may also be provided and connected to device 1450 through expansion interface 1472, which may include, for example, a SIMM (Single In Line Memory Module) card interface.
  • SIMM Single In Line Memory Module
  • expansion memory 1474 may provide extra storage space for device 1450, or may also store applications or other information for device 1450.
  • expansion memory 1474 may include instructions to carry out or supplement the processes described herein, and may include secure information also.
  • expansion memory 1474 may be provide as a security module for device 1450, and may be programmed with instructions that permit secure use of device 1450.
  • secure applications may be provided via the SIMM cards, along with additional information, such as placing identifying information on the SIMM card in a non-hackable manner.
  • the memory may include, for example, flash memory and/or NVRAM memory, as discussed below.
  • a computer program product is tangibly embodied in an information carrier.
  • the computer program product contains instructions that, when executed, perform one or more methods, such as those described herein.
  • the information carrier is a computer- or machine-readable medium, such as the memory 1464, expansion memory 1474, memory on processor 1452, or a propagated signal that may be received, for example, over transceiver 1468 or external interface 1462.
  • Device 1450 may communicate wirelessly through communication interface 1466, which may include digital signal processing circuitry where necessary. Communication interface 1466 may provide for communications under various modes or protocols, such as GSM voice calls, SMS, EMS, or MMS messaging, CDMA, TDMA, PDC, WCDMA, CDMA2000, or GPRS, among others. Such communication may occur, for example, through radio-frequency transceiver 1468. In addition, short-range communication may occur, such as using a Bluetooth, WiFi, or other such transceiver (not shown). In addition, GPS (Global Positioning System) receiver module 1470 may provide additional navigation- and location-related wireless data to device 1450, which may be used as appropriate by applications running on device 1450.
  • GPS Global Positioning System
  • Device 1450 may also communicate audibly using audio codec 1460, which may receive spoken information from a user and convert it to usable digital information. Audio codec 1460 may likewise generate audible sound for a user, such as through a speaker, e.g ., in a handset of device 1450. Such sound may include sound from voice telephone calls, may include recorded sound ( e.g ., voice messages, music files, etc.) and may also include sound generated by applications operating on device 1450.
  • Audio codec 1460 may receive spoken information from a user and convert it to usable digital information. Audio codec 1460 may likewise generate audible sound for a user, such as through a speaker, e.g ., in a handset of device 1450. Such sound may include sound from voice telephone calls, may include recorded sound ( e.g ., voice messages, music files, etc.) and may also include sound generated by applications operating on device 1450.
  • the computing device 1450 may be implemented in a number of different forms, as shown in the figure. For example, it may be implemented as a cellular telephone 1480. It may also be implemented as part of a smartphone 1482, personal digital assistant, or other similar mobile device.
  • implementations of the systems and techniques described herein can be realized in digital electronic circuitry, integrated circuitry, specially designed ASICs (application specific integrated circuits), computer hardware, firmware, software, and/or combinations thereof.
  • ASICs application specific integrated circuits
  • These various implementations can include implementation in one or more computer programs that are executable and/or interpretable on a programmable system including at least one programmable processor, which may be special or general purpose, coupled to receive data and instructions from, and to transmit data and instructions to, a storage system, at least one input device, and at least one output device.
  • the systems and techniques described herein can be implemented on a computer having a display device (e.g., a CRT (cathode ray tube) or LCD (liquid crystal display) monitor) for displaying information to the user and a keyboard and a pointing device (e.g., a mouse or a trackball) by which the user can provide input to the computer.
  • a display device e.g., a CRT (cathode ray tube) or LCD (liquid crystal display) monitor
  • a keyboard and a pointing device e.g., a mouse or a trackball
  • Other kinds of devices can be used to provide for interaction with a user as well; for example, feedback provided to the user can be any form of sensory feedback (e.g ., visual feedback, auditory feedback, or tactile feedback); and input from the user can be received in any form, including acoustic, speech, or tactile input.
  • the systems and techniques described herein can be implemented in a computing system that includes a back end component (e.g ., as a data server), or that includes a middleware component (e.g., an application server), or that includes a front end component (e.g., a client computer having a graphical user interface or a Web browser through which a user can interact with an implementation of the systems and techniques described herein), or any combination of such back end, middleware, or front end components.
  • the components of the system can be interconnected by any form or medium of digital data communication (e.g ., a communication network). Examples of communication networks include a local area network ("LAN”), a wide area network (“WAN”), and the Internet.
  • LAN local area network
  • WAN wide area network
  • the Internet the global information network
  • the computing system can include clients and servers.
  • a client and server are generally remote from each other and typically interact through a communication network.
  • the relationship of client and server arises by virtue of computer programs running on the respective computers and having a client-server relationship to each other.
  • a computing system provided herein can be configured to include one or more sample analyzers.
  • a sample analyzer can be configured to produce a plurality of signals about genomic DNA of at least one pair of human chromosomes of a cancer cell.
  • a sample analyzer can produce signals that are capable of being interpreted in a manner that identifies the genotype of loci along a chromosome.
  • a sample analyzer can be configured to carry out one or more steps of a SNP array-based assay or sequencing-based assay and can be configured to produce and/or capture signals from such assays.
  • a computing system provided herein can be configured to include a computing device.
  • the computing device can be configured to receive signals from a sample analyzer.
  • the computing device can include computer-executable instructions or a computer program (e.g ., software) containing computer-executable instructions for carrying out one or more of the methods or steps described herein.
  • such computer-executable instructions can instruct a computing device to analyze signals from a sample analyzer, from another computing device, from a SNP array-based assay, or from a sequencing-based assay.
  • the analysis of such signals can be carried out to determine genotypes, homozygosity or other chromosomal aberration s at certain loci, regions of CA, the number of CA Regions, to determine the size of CA Regions, to determine the number of CA Regions having a particular size or range of sizes, to determine whether or not a sample is positive for an HRD signature, to determine the number of Indicator CA Regions in at least one pair of human chromosomes, to determine a likelihood of a deficiency in BRCA1 and/or BRCA2 genes, to determine a likelihood of a deficiency in HDR, to determine a likelihood that a cancer patient will respond to a particular cancer treatment regimen (e.g., a regimen that includes a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, a PARP inhibitor, or a combination thereof), or to determine a combination of these items.
  • a particular cancer treatment regimen e.g., a regimen that includes a DNA
  • a computing system can include computer-executable instructions or a computer program (e.g ., software) containing computer-executable instructions for formatting an output providing an indication about the number of CA Regions, the size of CA Regions, the number of CA Regions having a particular size or range of sizes, whether or not a sample is positive for an HRD signature, the number of Indicator CA Regions in at least one pair of human chromosomes, a likelihood of a deficiency in BRCA1 and/or BRCA2 genes, to determine a likelihood of a deficiency in HDR, a likelihood that a cancer patient will respond to a particular cancer treatment regimen (e.g., a regimen that includes a DNA damaging agent, an anthracycline, a topoisomerase I inhibitor, radiation, a PARP inhibitor, or a combination thereof), or a combination of these items.
  • a particular cancer treatment regimen e.g., a regimen that includes a DNA damaging agent, an anthracycline,
  • a computing system can include computer-executable instructions or a computer program (e.g ., software) containing computer-executable instructions for determining a desired cancer treatment regimen for a particular patient based at least in part on the presence or absence of an HRD signature or on the number of Indicator CA Regions.
  • a computer program e.g ., software
  • a computing system can include a pre-processing device configured to process a sample (e.g ., cancer cells) such that a SNP array-based assay or sequencing-based assay can be performed.
  • a sample e.g ., cancer cells
  • pre-processing devices include, without limitation, devices configured to enrich cell populations for cancer cells as opposed to non-cancer cells, devices configured to lyse cells and/or extract genomic nucleic acid, and devices configured to enrich a sample for particular genomic DNA fragments.
  • kits for assessing samples e.g ., cancer cells
  • samples e.g ., cancer cells
  • this document provides kits for assessing cancer cells for the presence of an HRD signature or to determine the number of Indicator CA Regions in at least one pair of human chromosomes.
  • a kit provided herein can include either SNP probes (e.g ., an array of SNP probes for carrying out a SNP array-based assay described herein) or primers (e.g., primers designed for sequencing SNP regions via a sequencing-based assay) in combination with a computer program product containing computer-executable instructions for carrying out one or more of the methods or steps described herein (e.g., computer-executable instructions for determining the number of Indicator CA Regions).
  • a kit provided herein can include at least 500, 1000, 10,000, 25,000, or 50,000 SNP probes capable of hybridizing to polymorphic regions of human genomic DNA.
  • a kit provided herein can include at least 500, 1000, 10,000, 25,000, or 50,000 primers capable of sequencing polymorphic regions of human genomic DNA.
  • a kit provided herein can include one or more other ingredients for performing a SNP array-based assay or a sequencing-based assay. Examples of such other ingredients include, without limitation, buffers, sequencing nucleotides, enzymes ( e.g ., polymerases), etc. This document also provides the use of any appropriate number of the materials provided herein in the manufacture of a kit for carrying out one or more of the methods or steps described herein.
  • this document provides the use of a collection of SNP probes (e.g., a collection of 10,000 to 100,000 SNP probes) and a computer program product provided herein in the manufacture of a kit for assessing cancer cells for the presence of an HRD signature.
  • a collection of SNP probes e.g., a collection of 10,000 to 100,000 SNP probes
  • a computer program product provided herein in the manufacture of a kit for assessing cancer cells for the presence of an HRD signature.
  • the sample used is a frozen tumor sample.
  • the sample is from a particular breast cancer subtype chosen from triple negative, ER+/HER2-, ER-/HER2+, or ER+/HER2+.
  • the laboratory assay portion of the method, system, etc. comprises assaying the sample to sequence the BRCA1 and/or BRCA2 genes (as well as any other gene or genes in Table 1). In some embodiments, the laboratory assay portion of the method, system, etc.
  • the SNP analysis comprises assaying the sample to determine the allele dosage (e.g., genotype, copy number, etc.) for at least 10,000, 20,000, 30,000, 40,000, 50,000, 60,000, 70,000, 80,000, 90,000, 100,000 or more selected SNPs across the complete genome.
  • the SNP analysis is done using an oligonucleotide microarray as discussed above.
  • the BRCA sequence analysis, the SNP analysis, or both are performed using a probe capture (e.g., probes to each SNP to be analyzed and/or probes to capture the entire coding region of BRCA1 and/or BRCA2) with subsequent PCR enrichment technique (e.g., AgilentTM SureSelect XT).
  • the BRCA sequence analysis, the SNP analysis, or both are performed by processing the output from the enrichment technique using a "next-generation" sequencing platform (e.g., IlluminaTM HiSeq2500).
  • a "next-generation" sequencing platform e.g., IlluminaTM HiSeq2500.
  • the sample is analyzed for BRCA1/2 somatic and/or germline mutations, which may include large rearrangements.
  • the sample is analyzed for BRCA1 promoter methylation (e.g., by a qPCR assay (e.g., SA Biosciences)).
  • a sample is determined to have high methylation (or are "methylated") if the sample has greater than 10% (or 5%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%) methylation (e.g., % of BRCA1 or BRCA2 promoter CpGs methylated).
  • DNA from a patient's matched normal (non-tumor) tissue may be analyzed, e.g., to determine whether BRCA1 or BRCA2 mutations are germline or somatic.
  • LOH Region Score can be calculated by counting the number of LOH regions that are >15 Mb in length, but shorter than the length of a complete chromosome.
  • TAI Region Score can be calculated by counting the number of telomeric regions > 11 Mb in length with allelic imbalance that extends to one of the subtelomeres, but does not cross the centromere.
  • LST Region Score can be calculated by counting the number of breakpoints between regions longer than 10 megabases having stable copy number after filtering out regions shorter than 3 megabases.
  • BRCA1/2 deficiency can be defined as loss of function resulting from a BRCA1 or BRCA2 mutation, or methylation of the BRCA1 or BRCA2 promoter region, together with LOH in the affected gene.
  • response to treatment can be partial complete response ("pCR"), which in some embodiments can be defined as Miller-Payne 5 status following treatment (e.g., neoadjuvant).
  • the claimed method predicts BRCA deficiency with a p-value of at least 8 ⁇ 10 -12 6 ⁇ 10 -6 , 0.0009, 0.01, 0.03, 2 ⁇ 10 -16 , 3 ⁇ 10 -6 , 10 -6 , 0.0009, 8 ⁇ 10 -12 , 2 ⁇ 10 -16 , 8 ⁇ 10 -8 , 6 ⁇ 10 -6 , 3 ⁇ 10 -6 , or 0.0002 (e.g., each CA Region Score is predefined and optionally multiple scores are combined in such a way as to yield these p-values).
  • p-values are calculated according to Kolmogorov-Smirnov test.
  • HRD scores and age at diagnosis can be coded as a numeric (e.g., integer) variable
  • breast cancer stage and subtype can be coded as categorical variables
  • grade can be analyzed as either a numeric or categorical variable, or both.
  • p-values are two-sided.
  • logistic regression analysis can be used to predict BRCA1/2 deficiency based on an HRD score as disclosed herein, including the HRD-combined score).
  • BRCA deficiency status and HRD status can be combined to predict therapy response.
  • the disclosure can include a method of predicting patient (e.g., triple negative breast cancer patient) response to a cancer treatment regimen comprising a DNA damaging agent (e.g., platinum agent, e.g., cisplatin), an anthracycline, a topoisomerase I inhibitor, radiation, and/or a PARP inhibitor, the method comprising:
  • Example 1 LOH and TAI Region Scores Across Breast Cancer Subtypes and Association with BRCA1/2 Deficiency
  • An LOH signature based on whole genome tumor LOH profiles has been developed that is highly correlated with defects in BRCA1/2 and other HDR pathway genes in ovarian cancer ( Abkevich, et al., Patterns of Genomic Loss of Heterozygosity Predict Homologous Recombination Repair Defects, BR. J. CANCER (2012 )), and which predicts response to DNA-damaging agent (e.g., platinum-based neoadjuvant) therapy in breast cancer ( Telli et al., Homologous Recombination Deficiency (HRD) score predicts response following neoadjuvant platinum-based therapy in triple-negative and BRCA1/2 mutation-associated breast cancer (BC), CANCER RES.
  • DNA-damaging agent e.g., platinum-based neoadjuvant
  • HRD Homologous Recombination Deficiency
  • Frozen tumors were purchased from 3 commercial tissue biobanks. Approximately 50 randomly ascertained tumors from each of 4 breast cancer subtypes (triple negative, ER+/HER2-, ER-/HER2+, ER+/HER2+) were selected for analysis. A targeted custom hybridization panel was developed targeting BRCA1, BRCA2, and 50,000 selected SNPs across the complete genome. This panel, in combination with sequencing on the Illumina HiSeq2500, was used to analyze the tumors for BRCA1/2 somatic and germline mutations, including large rearrangements, and SNP allele dosages. BRCA1 promoter methylation was determined by a qPCR assay (SA Biosciences). When available, DNA from normal tissue was used to determine whether deleterious mutations were germline or somatic.
  • the SNP data was analyzed using an algorithm that determines the most likely allele specific copy number at each SNP location.
  • the LOH Region Score was calculated by counting the number of LOH regions that are > 15 Mb in length, but shorter than the length of a complete chromosome.
  • the TAI Region Score was calculated by counting the number of telomeric regions with allelic imbalance that are >11 Mb in length, but do not cross the centromere. Samples with low quality SNP data and/or with high contamination with normal DNA were excluded. 191 out of 213 samples yielded robust scores. Table 2: BRCA1/2 deficiency in breast cancer IHC subtypes.
  • Figure 5 shows LOH and TAI Region Scores across breast cancer IHC subtypes.
  • 5A LOH score; 5B: TAI score. Blue bars: BRCA1/2 deficient samples. Red bars: BRCA1/2 intact samples.
  • Elevated LOH and TAI Region Scores are each highly associated with BRCA1/2 deficiency in all subtypes of breast cancer; LOH and TAI Region Scores are highly significantly correlated; a Combined CA Region Score (i.e., combining LOH and TAI) shows the optimal correlation with BRCA1/2 deficiency in this dataset.
  • the combination of LOH-HRD and TAI-HRD scores can, based on the present disclosure, predict response to DNA-damaging and other agents (e.g. , platinum therapy) in triple negative breast cancer, and enable expansion of platinum use to other breast cancer subtypes.
  • Example 2 LOH, TAI, and LST Region Scores Across Breast Cancer Subtypes and Association with BRCA1/2 Deficiency
  • LST score was defined as the number of breakpoints between regions longer than 10 megabases having stable copy number after filtering out regions shorter than 3 megabases.
  • Example 3 Arithmetic mean of LOH, TAI, and LST Region Scores Across Breast Cancer Subtypes and Association with BRCA1/2 Deficiency
  • Invasive breast tumor samples and matched normal tissue were obtained from three commercial vendors. The samples were selected to give approximately equal numbers of all subtypes of breast cancer as defined by IHC analysis of ER, PR, and HER2. BRCA1 promoter methylation analysis was performed by qPCR. BRCA1/2 mutation screening and genome wide SNP profiles were generated using a custom Agilent SureSelect XT capture followed by sequencing on Illumina HiSeq2500. These data were used to calculate HRD-LOH, HRD-TAI, and HRD-LST scores.
  • SNP microarray data and clinical data were downloaded from a public repository for the cisplatin-1 and cisplatin-2 trial cohorts.
  • BRCA1/2 mutation data was not available for one of these cohorts.
  • All three HRD scores were calculated using publically available data, and analyzed for association with response to cisplatin. The two cohorts were combined to improve power.
  • HRD-LOH was calculated by counting the number of LOH regions >15 Mb in length, but shorter than the length of a complete chromosome.
  • HRD-TAI score was calculated by counting the number of regions > 11 Mb in length with allelic imbalance that extend to one of the subtelomeres, but do not cross the centromere.
  • HRD-LST score was the number of break points between regions longer than 10 Mb after filtering out regions shorter than 3 Mb.
  • the combined score was the arithmetic mean of the LOH/TAI/LST scores. All p values were from logistic regression models with BRCA deficiency or response to cisplatin as the dependent variable.
  • Table 6 shows BRCA1/2 mutation and BRCA1 promoter methylation frequency across four breast cancer subtypes.
  • BRCA1/2 variant analysis was successful on 100% of samples, while large rearrangement analysis was less robust with 198/214 samples producing data that passed QC metrics.
  • Deleterious mutations were observed in 24/214 individuals (one had a somatic mutation in BRCA1 and a germline mutation in BRCA2).
  • Matched normal DNA was available for 23/24 mutants, and was used to determine whether the identified mutation was germline or somatic.
  • BRCA1 promoter methylation analysis was successful on 100% of samples.
  • Figure 9 illustrates HRD scores in BRCA1/2 deficient samples.
  • Table 7 shows the association between the three HRD scores and BRCA 1/2 deficiency in the all-comers breast cohort. The combined score was the arithmetic mean of the three HRD scores.
  • Table 7 Breast Cancer Subtype All TNBC ER+/ HER- ER-/ HER2+ ER+/ HER2+ Number of Individuals 197 52 50 35 60 Number of BRCA1/2 Deficient (%) 38 (100) 23 (61) 5 (13) 3 (8) 7 (18) HRD-LOH mean BRCA1/2 Intact 7.2 8.2 7.1 8.3 6.0 BRCA1/2 Deficient 16.5 17.7 17.2 12.0 14.1 p value 1.3x10 -17 1.5x10 -8 0.0025 0.18 2.1x10 -5 HRD-TAI mean BRCA1/2 Intact 5.4 6.8 4.3 6.4 5.1 BRCA1/2 Deficient 13.7 13.5 15.0 7.7 15.9 p value 1.5x10 -19 2.2x10 -7 1.3x10 -5 0.58 1.4x10 -6 HRD-LST BRCA1/2 -7.0
  • Table 8 shows the association between HRD scores and pCR (Miller-Payne 5) in TNBC treated with cisplatin in a neoadjuvant setting. Data was available from samples from the Cisplatin-1 ( Silver et al., Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J. CLIN. ONCOL. 28:1145-53 (2010 )) and Cisplatin-2 (Birkbak et al., (2012)) trials. pCR was defined as those patients with Miller-Payne 5 status following neoadjuvant treatment. HRD-combined was the arithmetic mean of the three HRD scores.
  • the analyses in this Example 4 include the same 197 patient samples described in previous Examples. Briefly, 215 breast tumor samples were purchased as fresh frozen specimens from 3 commercial vendors. Samples were selected to give approximately equal representation of breast cancer subtypes according to IHC analysis of ER, PR, and HER2. 198 samples produced reliable HRD scores according to a Kolmogorov-Smirnov quality metric. One patient with a passing HRD score was removed from analysis due to unusual breast cancer subtype (ER/PR+ HER2-). Patient tumor and clinical characteristics are detailed in Table 9.
  • BRCA1/2 mutation screening and genome wide SNP profiles were generated using a custom Agilent SureSelect XT capture followed by sequencing on Illumina HiSeq2500. Methylation of the BRCA-1 promoter region was determined by qPCR. Samples with greater than 10% methylation were classified as methylated.
  • HRD scores were calculated from whole genome tumor loss of heterozygosity (LOH) profiles (HRD-LOH), telomeric allelic imbalance (HRD-TAI), and large-scale state transitions (HRD-LST), the three HRD scores combined in the "HRD-combined score" discussed in this Example 4.
  • LH whole genome tumor loss of heterozygosity
  • HRD-TAI telomeric allelic imbalance
  • HRD-LST large-scale state transitions
  • BRCA1/2 deficiency was defined as loss of function resulting from a BRCA-1 or BRCA-2 mutation, or methylation of the BRCA-1 promoter region, together with loss of heterozygosity (LOH) in the affected gene.
  • LHO heterozygosity
  • HRD scores and age at diagnosis were coded as numeric variable.
  • Breast cancer stage and subtype were coded as categorical variables.
  • Grade was analyzed as both a numeric and categorical variable, but was categorical unless otherwise noted. Coding grade as numerical is not appropriate unless the increased odds of BRCA1/2 deficiency is the same when comparing grade 2 to grade 1 patients, as when comparing grade 3 to grade 2 patients.
  • P-values reported for univariate logistic regression models are based on the partial likelihood ratio. Multivariate p-values are based on the partial likelihood ratio for change in deviance from a full model (which includes all relevant predictor) versus a reduced model (which includes all predictors except for the predictor being evaluated, and any interaction terms involving the predictor being evaluated). Odds ratios for HRD scores are reported per interquartile range.
  • Table 10 illustrates results from a 3-term multivariate logistic regression model with HRD-LOH, HRD-TAI, and HRD-LST as predictors of BRCA1/2 deficiency.
  • the HRD-combined score was finally compared to a model-based combined score which was optimized to predict BRCA1/2 deficiency in this patient set. While the HRD-combined score weights each of the HRD-LOH, HRD-TAI, and HRD-LST scores equally, the model-based score assigns the HRD-TAI score approximately twice the weight of the HRD-LOH or HRD-LST scores.
  • Table 11 shows results from univariate logistic regression. Odds ratios for HRD scores are reported per IQR of the score. The odds ratio for age is reported per year. The odds ratio for grade (numerical) is per unit.
  • Table 12 shows results from multivariate logistic regression. Odds ratios for HRD scores are reported per IQR of the score. The odds ratio for age is reported per year.
  • Example 4 the frequency of BRCA1/2 defects ranged from ⁇ 9 to ⁇ 16% across 4 subtypes of breast cancer as defined by IHC subtyping. Sequencing of matched tumor and normal DNA samples suggests that approximately 75% of the observed mutations were germline in origin. The primary method for loss of the second allele in breast cancer is via LOH, however ⁇ 24% of tumors also carried subsequent somatic deleterious mutations in the second allele. In addition, an apparently sporadic breast tumor was seen in one individual carrying a BRCA2 somatic deleterious mutation.
  • Liquid hybridization based target enrichment technologies have been developed for production of libraries for next generation sequencing. These methodologies enable targeted sequencing of regions of interest after reduction in genomic complexity, resulting in decreased sequencing costs. Preliminary tests indicated that the available assays are compatible with DNA derived from FFPE DNA.
  • this Example 4 we report the development of a capture panel which targets ⁇ 54,000 SNPs distributed across the genome. Allele counts from the sequencing information that this panel provides can be used for copy number and LOH reconstruction, and the calculation of all 3 of the HRD scores.
  • BRCA1 and BRCA2 capture probes may be included on the panel, as in this Example 4, which enable high quality mutation screening for deleterious variants in these genes in the same assay.
EP19216740.1A 2013-04-05 2014-04-04 Methoden und materialien zur beurteilung eines homologen rekombinationsfehlers Pending EP3693475A1 (de)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US201361809105P 2013-04-05 2013-04-05
US201361913762P 2013-12-09 2013-12-09
PCT/US2014/033014 WO2014165785A2 (en) 2013-04-05 2014-04-04 Methods and materials for assessing homologous recombination deficiency
EP14779403.6A EP2981624B1 (de) 2013-04-05 2014-04-04 Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
EP14779403.6A Division EP2981624B1 (de) 2013-04-05 2014-04-04 Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie

Publications (1)

Publication Number Publication Date
EP3693475A1 true EP3693475A1 (de) 2020-08-12

Family

ID=51659359

Family Applications (2)

Application Number Title Priority Date Filing Date
EP14779403.6A Active EP2981624B1 (de) 2013-04-05 2014-04-04 Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie
EP19216740.1A Pending EP3693475A1 (de) 2013-04-05 2014-04-04 Methoden und materialien zur beurteilung eines homologen rekombinationsfehlers

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP14779403.6A Active EP2981624B1 (de) 2013-04-05 2014-04-04 Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie

Country Status (10)

Country Link
US (3) US20140363521A1 (de)
EP (2) EP2981624B1 (de)
JP (4) JP6625045B2 (de)
AU (3) AU2014248007B2 (de)
CA (1) CA2908745C (de)
DK (1) DK2981624T3 (de)
ES (1) ES2777228T3 (de)
NZ (1) NZ712663A (de)
PT (1) PT2981624T (de)
WO (1) WO2014165785A2 (de)

Families Citing this family (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2609216T3 (en) 2010-08-24 2016-09-12 Dana Farber Cancer Inst Inc Methods to predict anti-cancer response
EP3225697A3 (de) 2010-12-30 2017-11-22 Foundation Medicine, Inc. Optimierung von multigenanalysen von tumorproben
WO2012174378A2 (en) 2011-06-17 2012-12-20 Myriad Genetics, Inc. Methods and materials for assessing allelic imbalance
CN104160037B (zh) 2011-12-21 2017-05-17 美瑞德生物工程公司 用于评估杂合性丢失的方法与材料
ES2687024T3 (es) 2012-02-23 2018-10-23 Children's Medical Center Corporation Procedimientos para predecir la respuesta contra el cáncer
EP2859118B1 (de) 2012-06-07 2017-11-22 Institut Curie Verfahren zur detektion der deaktivierung des pfades der homologen rekombination (brca1/2) in menschlichen tumoren
EP2981624B1 (de) * 2013-04-05 2019-12-18 Myriad Genetics, Inc. Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie
US11149316B2 (en) 2013-12-09 2021-10-19 Institut Curie Methods for detecting inactivation of the homologous recombination pathway (BRCA1/2) in human tumors
EA201691682A1 (ru) 2014-02-21 2017-02-28 Эпик Сайенсиз, Инк. Способы анализирования редких циркулирующих в крови клеток
EP3686288B1 (de) * 2014-08-15 2023-03-08 Myriad Genetics, Inc. Verfahren und materialien zur beurteilung eines mangels an homologer rekombination
JP6905934B2 (ja) * 2014-12-05 2021-07-21 ファウンデーション・メディシン・インコーポレイテッド 腫瘍試料の多重遺伝子分析
EP3230472A4 (de) * 2014-12-08 2018-06-13 Myriad Genetics, Inc. Verfahren und materialien zur vorhersage der reaktion auf niraparib
WO2016185406A1 (en) 2015-05-19 2016-11-24 Nadathur Estates Pvt. Ltd. Method for identification of a deficient brca1 function
WO2017079139A1 (en) 2015-11-03 2017-05-11 Epic Sciences, Inc. Single cell genomic profiling of circulating tumor cells (ctcs) in metastatic disease to characterize disease heterogeneity
WO2017106365A1 (en) * 2015-12-14 2017-06-22 Myriad Genetics, Inc. Methods for measuring mutation load
CA3010311A1 (en) 2016-01-06 2017-07-13 Epic Sciences, Inc. Single cell genomic profiling of circulating tumor cells (ctcs) in metastatic disease to characterize disease heterogeneity
WO2017165270A1 (en) * 2016-03-21 2017-09-28 Myriad Genetics, Inc. Homologous recombination deficiency to predict neoadjuvant chemotherapy necessity in bladder cancer
WO2017178509A1 (en) 2016-04-12 2017-10-19 Xentech Methods for predicting sensibility to treatment with parp inhibitors in cancerous patients
GB2555765A (en) 2016-05-01 2018-05-16 Genome Res Ltd Method of detecting a mutational signature in a sample
CA3021742A1 (en) * 2016-05-01 2017-11-09 Genome Research Limited Method of characterising a dna sample
RS65180B1 (sr) * 2016-06-29 2024-03-29 Tesaro Inc Postupci lečenja kancera jajnika
WO2020137076A1 (ja) * 2018-12-28 2020-07-02 国立大学法人 東京大学 Parp阻害剤に対する癌の感受性の予測方法および相同組換修復不全を有する癌の検出方法
CA3129831A1 (en) 2019-02-12 2020-08-20 Tempus Labs, Inc. An integrated machine-learning framework to estimate homologous recombination deficiency
CA3160566A1 (en) 2019-12-10 2021-06-17 Tempus Labs, Inc. Systems and methods for predicting homologous recombination deficiency status of a specimen
US20230018079A1 (en) * 2019-12-16 2023-01-19 Agilent Technologies, Inc. Genomic scarring assays and related methods
EP3945525A1 (de) 2020-07-27 2022-02-02 Sophia Genetics S.A. Verfahren zur identifizierung von chromosomaler räumlicher instabilität wie etwa homologem reparaturmangel in der next-generation-sequenzierung von daten mit niedriger coverage
CN111883211B (zh) * 2020-08-07 2021-04-23 张哲� 一种表征hrd同源重组修复缺陷的基因瘢痕及鉴定方法
US11718869B2 (en) * 2020-08-12 2023-08-08 Zhenyue Biotechnology Jiangsu Co., Ltd. Method and kit for determining genome instability based on next generation sequencing (NGS)
CN112410423B (zh) * 2020-11-03 2021-08-13 南京世和基因生物技术股份有限公司 同源重组缺失的标志物、检测方法以及检测系统
BE1029144B1 (fr) 2021-02-25 2022-09-20 Oncodna Méthode de caracterisation d'une tumeur à l'aide d'un séquençage ciblé
CN112820351A (zh) * 2021-03-01 2021-05-18 江苏医联生物科技有限公司 检测肿瘤患者的突变和hrd评分指导用药的方法
CN117561572A (zh) * 2021-06-25 2024-02-13 基金会医学公司 对同源修复缺陷进行分类的系统和方法
WO2023107509A1 (en) * 2021-12-08 2023-06-15 Myriad Genetics, Inc. Methods and materials for assessing homologous recombination deficiency in breast cancer subtypes
KR102523938B1 (ko) * 2022-02-04 2023-04-19 가톨릭대학교 산학협력단 상동 재조합 결핍증 예측 모델의 모델링 방법
WO2024083971A1 (en) 2022-10-19 2024-04-25 Vib Vzw Method of determining loss of heterozygosity status of a tumor

Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3590028A (en) 1967-04-18 1971-06-29 Farmaceutici Italia Adriamycin derivatives
US3892790A (en) 1972-04-10 1975-07-01 Rustenburg Platinum Mines Ltd Compositions containing platinum
US3904663A (en) 1972-04-10 1975-09-09 Rustenburg Platinum Mines Ltd Compositions containing platinum
US4138480A (en) 1976-01-16 1979-02-06 Gosalvez Mario Novel anthracycline glycosides and methods of preparing the same
US4950738A (en) 1984-09-13 1990-08-21 Cytogen Corporation Amine derivatives of anthracycline antibiotics
US5177075A (en) 1988-08-19 1993-01-05 Warner-Lambert Company Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process
US5295944A (en) 1991-05-14 1994-03-22 Dana-Farber Cancer Institute Method for treating a tumor with ionizing radiation
US5633016A (en) 1991-11-15 1997-05-27 Smithkline Beecham Corporation Combination chemotherapy
US6087340A (en) 1985-08-02 2000-07-11 Pharmacia & Upjohn, Inc. Injectable ready-to-use solutions containing an antitumor anthracycline glycoside
US6403563B1 (en) 1998-03-24 2002-06-11 Pharmacia & Upjohn S.P.A. Antitumor composition containing a synergistic combination of an anthracycline derivative with a camptothecin derivate
US7351701B2 (en) 2003-07-25 2008-04-01 Cancer Research Technology Limited Therapeutic compounds
US7485707B2 (en) 2003-07-02 2009-02-03 Solux Corporation Thermally stable crystalline epirubicin hydrochloride and method of making the same
KR100925337B1 (ko) * 2009-05-25 2009-11-09 주식회사 마크로젠 염색체 수 이상 검출을 통한 유방암 약물 반응성 예측
US7754684B2 (en) 2003-06-11 2010-07-13 Access Pharmaceuticals, Inc. Macromolecular platinum chelates
US7759510B2 (en) 2003-08-13 2010-07-20 University Of South Florida Platinum complexes and methods of use
US7759488B2 (en) 2005-06-30 2010-07-20 Bionumerik Pharmaceuticals, Inc. Monoazole ligand platinum analogs
US7868040B2 (en) 2001-09-03 2011-01-11 Cancer Research Technology Limited Anti-cancer combinations
US7915280B2 (en) 2000-12-01 2011-03-29 Eisai Inc. Compounds and their uses
WO2011160063A2 (en) 2010-06-18 2011-12-22 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
WO2012027224A1 (en) 2010-08-24 2012-03-01 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
WO2013130347A1 (en) * 2012-02-23 2013-09-06 The Children's Hospital Corporation Methods for predicting anti-cancer response

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2981624B1 (de) * 2013-04-05 2019-12-18 Myriad Genetics, Inc. Verfahren zur beurteilung eines mangels an homologer rekombination und zur vorhersage der reaktion auf eine krebstherapie

Patent Citations (21)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3590028A (en) 1967-04-18 1971-06-29 Farmaceutici Italia Adriamycin derivatives
US3892790A (en) 1972-04-10 1975-07-01 Rustenburg Platinum Mines Ltd Compositions containing platinum
US3904663A (en) 1972-04-10 1975-09-09 Rustenburg Platinum Mines Ltd Compositions containing platinum
US4138480A (en) 1976-01-16 1979-02-06 Gosalvez Mario Novel anthracycline glycosides and methods of preparing the same
US4950738A (en) 1984-09-13 1990-08-21 Cytogen Corporation Amine derivatives of anthracycline antibiotics
US6087340A (en) 1985-08-02 2000-07-11 Pharmacia & Upjohn, Inc. Injectable ready-to-use solutions containing an antitumor anthracycline glycoside
US5177075A (en) 1988-08-19 1993-01-05 Warner-Lambert Company Substituted dihydroisoquinolinones and related compounds as potentiators of the lethal effects of radiation and certain chemotherapeutic agents; selected compounds, analogs and process
US5295944A (en) 1991-05-14 1994-03-22 Dana-Farber Cancer Institute Method for treating a tumor with ionizing radiation
US5633016A (en) 1991-11-15 1997-05-27 Smithkline Beecham Corporation Combination chemotherapy
US6403563B1 (en) 1998-03-24 2002-06-11 Pharmacia & Upjohn S.P.A. Antitumor composition containing a synergistic combination of an anthracycline derivative with a camptothecin derivate
US7915280B2 (en) 2000-12-01 2011-03-29 Eisai Inc. Compounds and their uses
US7868040B2 (en) 2001-09-03 2011-01-11 Cancer Research Technology Limited Anti-cancer combinations
US7754684B2 (en) 2003-06-11 2010-07-13 Access Pharmaceuticals, Inc. Macromolecular platinum chelates
US7485707B2 (en) 2003-07-02 2009-02-03 Solux Corporation Thermally stable crystalline epirubicin hydrochloride and method of making the same
US7351701B2 (en) 2003-07-25 2008-04-01 Cancer Research Technology Limited Therapeutic compounds
US7759510B2 (en) 2003-08-13 2010-07-20 University Of South Florida Platinum complexes and methods of use
US7759488B2 (en) 2005-06-30 2010-07-20 Bionumerik Pharmaceuticals, Inc. Monoazole ligand platinum analogs
KR100925337B1 (ko) * 2009-05-25 2009-11-09 주식회사 마크로젠 염색체 수 이상 검출을 통한 유방암 약물 반응성 예측
WO2011160063A2 (en) 2010-06-18 2011-12-22 Myriad Genetics, Inc. Methods and materials for assessing loss of heterozygosity
WO2012027224A1 (en) 2010-08-24 2012-03-01 Dana-Farber Cancer Institute, Inc. Methods for predicting anti-cancer response
WO2013130347A1 (en) * 2012-02-23 2013-09-06 The Children's Hospital Corporation Methods for predicting anti-cancer response

Non-Patent Citations (14)

* Cited by examiner, † Cited by third party
Title
ABKEVICH ET AL.: "Patterns of Genomic Loss of Heterozygosity Predict Homologous Recombination Repair Defects", BR. J. CANCER, 2012
ANONYMOUS: "Myriad's HRD Test Significantly Predicts Response to Cisplatin Treatment in Patients With Triple Negative Breast Cancer in Second Research Study", 14 December 2013 (2013-12-14), XP055312045, Retrieved from the Internet <URL:http://files.shareholder.com/downloads/MYGN/3039924177x0x713818/74F0EC3E-B337-4724-A51B-63BA3BD75A08/MYGN_News_2013_12_14_General.pdf> [retrieved on 20161019] *
BIRKBAK ET AL.: "Telomeric allelic imbalance indicates defective DNA repair and sensitivity to DNA-damaging agents", CANCER DISCOV., 2012
GORANSSON ET AL., PLOS ONE, vol. 4, no. 6, 2009, pages e6057
MARIEKE A VOLLEBERGH ET AL: "Genomic instability in breast and ovarian cancers: translation into clinical predictive biomarkers", CMLS CELLULAR AND MOLECULAR LIFE SCIENCES, BIRKHÄUSER-VERLAG, BA, vol. 69, no. 2, 16 September 2011 (2011-09-16), pages 223 - 245, XP019993251, ISSN: 1420-9071, DOI: 10.1007/S00018-011-0809-0 *
ML TELLI ET AL: "Abstract PD09-04: Homologous Recombination Deficiency (HRD) score predicts pathologic response following neoadjuvant platinum-based therapy in triple-negative and BRCA1/2 mutation-associated breast cancer (BC)", CANCER RESEARCH, 15 December 2012 (2012-12-15), XP055312264, Retrieved from the Internet <URL:http://cancerres.aacrjournals.org/content/72/24_Supplement/PD09-04> [retrieved on 20161019] *
N. J. BIRKBAK ET AL: "Telomeric Allelic Imbalance Indicates Defective DNA Repair and Sensitivity to DNA-Damaging Agents", CANCER DISCOVERY, vol. 2, no. 4, 22 March 2012 (2012-03-22), pages 366 - 375, XP055162779, ISSN: 2159-8274, DOI: 10.1158/2159-8290.CD-11-0206 *
NANNYA ET AL., CANCER RES., vol. 65, 2005, pages 6071 - 6079
O'BRIEN ET AL.: "Converting cancer mutations into therapeutic opportunities", EMBO MOL. MED., vol. 1, 2009, pages 297 - 299, XP055569693, DOI: 10.1002/emmm.200900044
POPOVA ET AL.: "Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation", CANCER RES., vol. 72, 2012, pages 5454 - 5462, XP055341888, DOI: 10.1158/0008-5472.CAN-12-1470
SILVER ET AL.: "Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer", J. CLIN. ONCOL., vol. 28, 2010, pages 1145 - 53, XP055010413, DOI: 10.1200/JCO.2009.22.4725
T. POPOVA ET AL: "Ploidy and large-scale genomic instability consistently identify basal-like breast carcinomas with BRCA1/2 inactivation", CANCER RESEARCH, 29 August 2012 (2012-08-29), XP055039519, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-12-1470 *
TELLI ET AL.: "Homologous Recombination Deficiency (HRD) score predicts response following neoadjuvant platinum-based therapy in triple-negative and BRCA1/2 mutation-associated breast cancer (BC)", CANCER RES., 2012
V ABKEVICH ET AL: "Patterns of genomic loss of heterozygosity predict homologous recombination repair defects in epithelial ovarian cancer", BRITISH JOURNAL OF CANCER, vol. 107, no. 10, 6 November 2012 (2012-11-06), pages 1776 - 1782, XP055085758, ISSN: 0007-0920, DOI: 10.1038/bjc.2012.451 *

Also Published As

Publication number Publication date
CA2908745A1 (en) 2014-10-09
NZ712663A (en) 2021-07-30
JP2021036903A (ja) 2021-03-11
EP2981624A2 (de) 2016-02-10
WO2014165785A2 (en) 2014-10-09
AU2020204028A1 (en) 2020-07-09
ES2777228T3 (es) 2020-08-04
AU2022211880A1 (en) 2022-09-01
US20180030544A1 (en) 2018-02-01
JP6625045B2 (ja) 2019-12-25
DK2981624T3 (da) 2020-03-02
EP2981624B1 (de) 2019-12-18
AU2020204028B2 (en) 2022-08-18
US20140363521A1 (en) 2014-12-11
JP2016523511A (ja) 2016-08-12
JP2023012488A (ja) 2023-01-25
AU2014248007A1 (en) 2015-10-15
PT2981624T (pt) 2020-03-23
EP2981624A4 (de) 2016-11-30
AU2014248007B2 (en) 2020-03-26
CA2908745C (en) 2023-03-14
JP2019165731A (ja) 2019-10-03
US20210310079A1 (en) 2021-10-07

Similar Documents

Publication Publication Date Title
US20210310079A1 (en) Methods and materials for assessing homologous recombination deficiency
US20230117133A1 (en) Methods and materials for assessing homologous recombination deficiency
AU2020223754B2 (en) Methods and materials for assessing loss of heterozygosity
EP3327148B1 (de) Verfahren zur vorherbestimmung des status der gene brca1 und brca2 in einer krebszelle
WO2017165270A1 (en) Homologous recombination deficiency to predict neoadjuvant chemotherapy necessity in bladder cancer
US20230212653A1 (en) Methods and materials for assessing homologous recombination deficiency in breast cancer subtypes

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION HAS BEEN PUBLISHED

AC Divisional application: reference to earlier application

Ref document number: 2981624

Country of ref document: EP

Kind code of ref document: P

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20210205

RBV Designated contracting states (corrected)

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR